The Gut Microbiome Bench
The Gut Microbiome Bench
The influence of the gut microbiome on human health and disease has been estab-
lished in recent years through advances in high-throughput DNA sequencing. The
Gut Microbiome: Bench to Table presents a scientific introduction to this topic
and analyzes the research on how the microbiome is affected by nutrients and how
dietary modifications can alter the microbiome.
The Gut Microbiome: Bench to Table is divided into three sections. The first sec-
tion details the current state of laboratory-scale analysis of gut microbiome samples
and how we can identify the communities and their functional repertoire. Section II
explains the next phase of translational research models such as preclinical animal
studies, proof of concept safety, and efficacy human trials. The third section demon-
strates the effectiveness of therapeutic treatments in larger populations. It addresses
how diet influences the gut microbiome and presents an array of approaches that
have been reported, including a discussion of issues of the safety of probiotics and
selected supplements and micronutrients.
This book is essential for clinicians, dietitians, and food and nutrition
professionals who wish to have the most up-to-date knowledge in understanding
gut microbiome and diet.
The Gut Microbiome
Bench to Table
Edited by
Vivian C.H. Wu
First edition published 2023
by CRC Press
6000 Broken Sound Parkway NW, Suite 300, Boca Raton, FL 33487-2742
Reasonable efforts have been made to publish reliable data and information, but the author and
publisher cannot assume responsibility for the validity of all materials or the consequences of
their use. The authors and publishers have attempted to trace the copyright holders of all material
reproduced in this publication and apologize to copyright holders if permission to publish in this
form has not been obtained. If any copyright material has not been acknowledged please write and
let us know so we may rectify in any future reprint.
Except as permitted under U.S. Copyright Law, no part of this book may be reprinted, reproduced,
transmitted, or utilized in any form by any electronic, mechanical, or other means, now known or
hereafter invented, including photocopying, microfilming, and recording, or in any information
storage or retrieval system, without written permission from the publishers.
For permission to photocopy or use material electronically from this work, access www.copyright.
com or contact the Copyright Clearance Center, Inc. (CCC), 222 Rosewood Drive, Danvers, MA
01923, 978-750-8400. For works that are not available on CCC please contact mpkbookspermissions@
tandf.co.uk
Trademark notice : Product or corporate names may be trademarks or registered trademarks and
are used only for identification and explanation without intent to infringe.
DOI: 10.1201/b22970
Typeset in Times
by codeMantra
Dedication
ix
x Preface
xi
Editor
Dr. Vivian C.H. Wu is the Research Leader of the Produce Safety and Microbiology
Research Unit, one of the six Research Units at the U.S. Dept. of Agriculture (USDA)-
Agriculture Research Service (ARS) Western Regional Research Center in Albany,
California. In 2021, she was the acting Associate Area Director of USDA-ARS
Pacific West Area (PWA), providing leadership and line management to the program
and staff in the PWA, which covers eight states with more than 2000 employees.
Before joining USDA ARS, Dr. Wu served as a Full Professor at the University of
Maine, where she led the Pathogenic Microbiology Laboratory. She currently serves
as an Adjunct Professor at Washington State University, Texas Tech University, and
National Chiao Tung University (Taiwan). Dr. Wu earned her Ph.D. in food science
with an emphasis on microbiology under Dr. Daniel Y. C. Fung from the Department
of Animal Sciences and Industry at Kansas State University. She was appointed to
the faculty at the University of Maine in 2003 and has developed an exceptionally
creative research program that led to attaining her international stature in micro-
bial food safety, pathogen detection, interventions, and functional foods. Since
August 2015, she has been serving as the Research Leader for the Produce Safety
and Microbiology Research Unit, USDA-ARS, where she oversees more than 60
scientists (PIs), technicians, support staff, and postdocs, in addition to student interns
and volunteers. Dr. Wu has published 145 peer-reviewed journal articles, four book
chapters, 18 proceedings, 161 peer-reviewed abstracts, one patent and six invention-
patent disclosures, and 51 technical reports (375 publications in total). Dr. Wu is a
fundholder of an $8M annual budget for three ARS CRIS research projects in addi-
tion to more than $18 million in external competitive grants she received for her
research program. Before ARS, she had garnered more than $12 million in external
competitive grants as the Principal or Co-Investigator to her research program dur-
ing her 11 years at the University of Maine. Additionally, Dr. Wu received the 2012
International Bimbo Pan-American Nutrition, Food Science and Technology Award
(out of 107 nominees) for her achievement in the development of biosensors, the
2011 Eastern Scholar Award from Shanghai Institutions of Higher Learning for her
research achievement, and the 2012 Distinguished Service Award from the Chinese
American Food Society for her service contribution to the society. She received
the Chinese American Food Society (CAFS) Lifetime achievement award in 2015
and CAFS Professional achievement award in 2020. She received National Chung-
Hsing University Distinguished Alumni Achievement Award in 2020. Her research
accomplishments have also been translated into successfully training undergraduate
and graduate students and postdoctoral research scientists, resulting in more than
28 prestigious national and international research awards. She has graduated seven
Ph.D. students, 24 M.S. thesis students, three undergraduate honors thesis students,
29 undergraduate research capstone students, and nine high school research capstone
students. She currently advises three postdocs and co-advises four Ph.D. students.
Dr. Wu’s internationally recognized expertise has resulted in invitations from the
USDA, FDA, universities, and research institutes in the U.S. and other countries
xiii
xiv Editor
to give talks about her research and insights on food safety. She gave 122 invited
presentations, including 54 international invitations, and made 135 presentations at
national and international meetings. She was the Spotlight Scientist selected by the
Center for Produce Safety at the University of California-Davis (2013), highlighting
her research achievement in produce safety. Dr. Wu was appointed to serve on the
Board of Agriculture to advise the Chancellor of the University of Maine System
and the President of the University of Maine on matters concerning agricultural
research and extension development. She received invitations to serve on prestigious
grant panels and ad hoc proposal reviews. Before ARS, She was invited to chair
an ARS review panel in the Quality and Utilization of Agricultural Crops National
Program. She is the Editor-in-Chief of the Journal of Food Safety. She serves on 16
international journal editorial boards. She served as the elected treasurer and several
standing committees of the Chinese American Food Society for ten years before
becoming the President of the Chinese American Food Society (CAFS), 2015-2016.
Contributors
Raphael D. Ayivi Hafize Fidan
Department of Food and Nutritional Department of Nutrition and Tourism
Sciences University of Food Technology
North Carolina A&T State University Plovdiv, Bulgaria
University of North Carolina
Greensboro, North Carolina Rabin Gyawali
Department of Nanoscience, Joint Department of Food and Nutritional
School of Nanoscience and Sciences
Nanoengineering North Carolina A&T State University
University of North Carolina Greensboro, North Carolina
Greensboro, North Carolina
Gianluca Ianiro
Reza V. Bakhshayesh Digestive Disease Center
Department of Food Biotechnology, Catholic University of Rome
Branch for Northwest & Rome, Italy
West Region, Agricultural
Research, Education and Salam A. Ibrahim
Extension Organization (AREEO) Department of Food and Nutritional
Agricultural Biotechnology Research Sciences
Institute of Iran North Carolina A&T State University
Tabriz, Iran Greensboro, North Carolina
Department of Food Science and
Technology Dorothy Klimis-Zacas
University of Tabriz School of Food and Agriculture
Tabriz, Iran University of Maine
Orono, Maine
Claudia Caglioti
Digestive Disease Center Aleksandra S. Kristo
Catholic University of Rome Department of Food Science and Nutrition
Rome, Italy California Polytechnic
State University
Yu-Chieh Cheng San Luis Obispo, California
Agricultural Biotechnology Research
Center Alison Lacombe
Academia Sinica Produce Safety and Microbiology
Taipei, Taiwan Research Unit, Agricultural
Biotechnology Center in Southern Research Service
Taiwan United States Department of
Academia Sinica Agriculture
Tainan, Taiwan Albany, California
xv
xvi Contributors
xvii
xviii Introduction
environment, and diet. In turn, the microbiome influences host metabolism, calorie
absorption, immune reactions, and behavior. The gut microbiome modulates its host
environment by releasing its own metabolites, modifying the metabolites of the host,
and synthesizing vitamins and enzymes.
Consumers are aware of the potential healing properties of dietary regimens and
foods and have fueled the demand for products that promote health. The desire for
food that meets nutritional needs and can improve health has led food scientists to
develop the concept of functional foods. Dietary supplements/nutrients offer new
tools for improving host health by either preventing or ameliorating the symptoms
of immune disorders, cancer, obesity, and other diseases. This book will discuss the
microbes used as probiotics and the best industrial practices for optimal use. One
future direction is optimizing the composition of infant formulas and other food
products in maintaining gut health.
As the concept of gut health continues to evolve, researchers investigate the poten-
tial benefits of enhancing beneficial species through diet or other interventions. There
are many possible sources of beneficial products for the gut, which can be found in
the natural environment or synthesized based on research indicating certain func-
tional groups with benefits. However, more work is needed to determine which prebi-
otic regimen is best for dietary recommendations, although recommendations cannot
probably be applied ubiquitously across the population. This opens a fascinating field
of inquiry into tailoring probiotic and prebiotic regimens for optimum health. Using
a translation approach with different models, research can begin to elucidate the
causal relationship between the diet, the host genetics, and the gut microbiome.
Section I
T0 Benchwork,
Including Techniques for
Microbiome Studies
1 Tools and Resources
Enabling Marker
Gene–Based
Microbiome Studies
Jianan Liu and Robert W. Li
United States Department of Agriculture
CONTENTS
Introduction ................................................................................................................4
Historical Perspective of Microbiome Studies...........................................................6
High-Throughput Sequencing Technologies .............................................................8
454 Pyrosequencing and Supported Oligonucleotide Ligation
and Detection ........................................................................................................8
Illumina Platform ..................................................................................................9
Ion Torrent ........................................................................................................... 10
Long Read Sequencing Platforms ....................................................................... 11
SSU Reference Databases ........................................................................................ 12
Greengenes .......................................................................................................... 12
SILVA ..................................................................................................................13
Ribosomal Database Project ............................................................................... 13
EzBioCloud ......................................................................................................... 14
UNITE ................................................................................................................. 14
PR2....................................................................................................................... 15
Marker Gene Sequence Analysis Pipelines.............................................................. 15
QIIME 1 and QIIME 2 ........................................................................................ 15
mothur ................................................................................................................. 18
FROGS ................................................................................................................ 19
iMAP ...................................................................................................................20
Recent Advances in Computational Tool Development........................................... 21
Uniqueness of Marker Gene Survey Datasets ..................................................... 21
Statistical Testing and Differentially Abundant Taxon Identification ................. 21
Balances and Microbial Signature Identification ................................................24
Tools for Network Analysis and Their Applications ...........................................26
DOI: 10.1201/b22970-2 3
4 The Gut Microbiome: Bench to Table
INTRODUCTION
The microbiome is referred to as “the entire habitat, including the microorgan-
isms (bacteria, archaea, lower and higher eukaryotes, and viruses), their genomes
(i.e., genes), and the surrounding environmental conditions” (Marchesi and Ravel,
2015). Trillions of microorganisms from archaea, bacteria, and eukarya and their
viruses colonize the gastrointestinal tract of humans and animals from birth and
play critical roles in their development and disease. It is still debatable whether the
placenta is sterile. Most studies show that the detection of the placental microbiome
is largely due to contamination (Kuperman et al., 2019; Theis et al., 2019). However,
a recent study of a human fetus microbiome revealed the possibility of the exposure
of the fetus to bacteria and their metabolites in utero (Stinson et al., 2019). Moreover,
questions have been raised about the widely quoted notion that the total number of
microbial cells outnumbers human cells by 10 to 1. The latest study suggests that the
estimated number of bacteria in the human body is 3.8 × 1013, approximately a 1:1
ratio with the number of human cells (Sender et al., 2016). Nevertheless, the micro-
biome plays vital functions in host physiology and nutrition, including modulating
the host immunity, protecting the host against invading pathogens, and affecting the
host organ development. It has long been known that microorganisms in the gastro-
intestinal tract act as a metabolic organ and are responsible for nutrient production
and utilization. For ruminant species, the microorganisms in the rumen and hindgut
convert carbohydrates (mainly plant fiber) into readily absorbable short-chain fatty
acids, which provide up to 75% of their total metabolizable energy (Li et al., 2012). In
monogastric species, such as humans and pigs, microbial fermentation in the hindgut
allows the host to harvest extra energy from otherwise indigestible carbohydrates.
Numerous gut bacteria, especially those from Prevotella, also participate in dietary
protein degradation, including oligopeptide degradation and deamination. In addi-
tion, gut microorganisms produce vitamins for the host (LeBlanc et al., 2013). As
a result, ruminants do not need a dietary supply of water-soluble vitamins, while
a significant portion of our daily vitamin requirement, particularly water-soluble
B vitamins and vitamin K, are synthesized by gut microbes. Gut microorganisms
also affect host organ development. Evidence accumulated from studies of germ-
free animals suggests the gut microbiome is essential for proper intestinal motility,
morphology, function, and epithelial renewal (Sommer and Bäckhed, 2013). Most
notably, the gut microbiome is instrumental in promoting the development of both
innate and adapted immune systems (Duerkop et al., 2009; Sansonetti and Di Santo,
2007). Indeed, the strong induction of innate immunity followed by the stimula-
tion of adaptive immune responses can be detected only four days after colonization
of germ-free mice with the total fecal microbial community (El Aidy et al., 2012).
Microbiome-derived metabolites, including short-chain fatty acids, directly regulate
innate and adaptive immune cells (Rooks and Garrett, 2016). However, disruption
Tools and Resources for Microbe Analysis 5
of the homeostasis of the host microbiome can result in severe pathological con-
sequences, such as chronic inflammation and metabolic dysfunction (Sommer and
Bäckhed, 2013). The gut microbiome has been linked to the development of obesity
and metabolic disorders, such as diabetes and insulin resistance (Ley, 2010). The
intestinal microbiome affects the pathogenesis of inflammatory bowel diseases, such
as Crohn’s disease and ulcerative colitis (Baker et al., 2009; Joossens et al., 2011;
Schwiertz et al., 2010). Inflammatory bowel disease patients tend to have an aberrant
microbiome, characterized by the depletion of commensal bacteria, notably members
of the phyla Firmicutes and Bacteroidetes. Furthermore, the altered microbiome has
been suggested to affect the carcinogenesis of several tumors, including colorectal
gastric tumors and cancerous liver tumors (Fox et al., 2010). Evidence also suggests
the gut microbiome affects viral infection (Wilks and Golovkina, 2012) and plays a
role in HIV progression (Ellis et al., 2011). As a result, the gut microbiome has been
touted as a therapeutic target or as a drug target (Cani and Delzenne, 2011).
While the structure and functions of the gut microbiome have been extensively
explored in the past few years, the complex interrelationships of the microbiome
within a host still require higher-resolution studies using comprehensive toolsets
to promote new discoveries (Heintz-Buschart and Wilmes, 2018). Because of the
complexity and functional importance of the microbiome, numerous methods and
bioinformatic tools have been developed in recent years to conduct comprehensive
microbiome studies using culture-independent approaches. For example, enzymes
that catalyze chemical reactions are encoded by genes present in microbial com-
munities from a broad spectrum of environmental conditions. Therefore, functional
screening has become an increasingly important tool to discover novel biomolecules
for applications in biotechnology and medicine. This approach, also termed func-
tional metagenomics, provides direct access to largely unexploited microbial genetic
diversity in the environment. In the meantime, the rapid advancement of ultra-low
cost and high-throughput or parallel sequencing technologies and the development
of bioinformatics tools and various databases have stimulated sequencing-based
microbiome studies, including whole genome shotgun–based computational metage-
nomics and marker gene–based microbiome studies. The former allows us to have a
holistic assessment of the functional capacity and metabolic potential of the micro-
bial community in its habitat.
Marker gene–based microbiome profiling is a rapid alternative to whole genome
shotgun–based approaches in quantifying the structure and composition of microbial
communities. Due to their functional constancy and highly conserved nature, small
subunits (SSUs) of the rRNA gene, especially the 16S rRNA gene, have been used
the most as phylogenetic markers for genus and species identification and taxonomic
classification of bacteria and archaea. The 16S rRNA gene is large enough (~1,500
nucleotides) compared to 5S rRNA genes to carry adequate information for com-
parisons and is small enough compared to 23S rRNA genes (~2,900 nucleotides)
to be conveniently analyzed. As a homologue of prokaryotic 16S rRNA genes, the
eukaryotic 18S rRNA gene is also widely used as a phylogenetic marker for fungi and
other eukaryotes. The presence of hypervariable (HV) regions in these genes reflects
the evolutionary divergence of microorganisms and can be explored to differentiate
even highly related species (Woese, 1987). The internal transcribed spacer (ITS),
6 The Gut Microbiome: Bench to Table
located between the 18S and 5.8S rRNA genes, is used as a marker specifically to
identify a broad range of fungi. The ITS is more variable than the 18S rRNA genes
and is therefore more appropriate as the genetic marker to study intraspecific genetic
diversity (Schoch et al., 2012). Furthermore, many protein-coding single-copy genes,
such as the RNA polymerase β subunit gene (rpoB), have also been explored as
molecular markers for microbial ecology studies (Case et al., 2007).
Many molecular methods have been developed in the past to characterize SSUs
of rRNA genes, such as polymerase chain reaction (PCR)–based cloning and tra-
ditional sequencing and culture-independent molecular approaches, including
denaturing gradient gel electrophoresis/temperature gradient gel electrophoresis,
PCR-restriction fragment length polymorphism (RFLP), and terminal restriction
fragment length polymorphism (TRFLP). Additionally, 16S probe-based fluorescent
in situ hybridization (FISH) is a powerful tool for the visualization and identification
of microorganisms in their native habitats (Crocetti et al., 2000; Ginige et al., 2004).
The recent renaissance of the microbial marker gene–based study to characterize the
gut microbiome is largely driven by rapid advances in next-generation DNA sequenc-
ing technologies (Gloor et al., 2010; Tringe and Hugenholtz, 2008) and the develop-
ment of related data analysis pipelines and databases (Balvočiūtė and Huson, 2017;
Caporaso et al., 2010). Moreover, several new concepts and computational tools are
in rapid development. In this chapter, we will focus on recent advances in marker
gene–based approaches and bioinformatics tools essential for microbiome studies.
(DeSantis et al., 2007). The advantage of this conventional cloning method is that
the clean sequence of the entire 16S rRNA gene can be obtained and then sequenced,
which provides a reliable and precise picture of prokaryotes in a microbial commu-
nity. However, the heavy workload makes the method extremely time-consuming,
laborious, and expensive (Green and Sambrook, 2012).
A variety of molecular methods were then developed to overcome the issues asso-
ciated with conventional cloning. Most of these methods utilize DNA hybridization
and DNA length and sequence polymorphisms to study microbial communities. In
1993, a method called denaturing gradient gel electrophoresis was applied to ana-
lyze the genetic diversity of complex prokaryotic communities (Muyzer et al., 1993).
Shortly after, a similar method called temperature gradient gel electrophoresis was
developed. Denaturing gradient gel electrophoresis examines microbial genetic pat-
terns via a chemical gradient to denature the PCR-amplified 16S molecules as they
move across an acrylamide gel, while temperature gradient gel electrophoresis is
based on melting point differences in DNA molecules with a linear temperature gra-
dient in the gel (Muyzer et al., 1993; Muyzer and Smalla, 1998). These two methods
are more rapid and reproducible than the conventional cloning method.
Two other efficient methods, PCR-RFLP and TRFLP, are based on RFLP. These
methods can be applied to 16S rRNA, ITS-5.8S rRNA-ITS2, or 23S rRNA genes
to characterize microbial diversity (Guillamön et al., 1998; Leaw et al., 2006).
For PCR-RFLP, PCR products amplified from SSU rRNA genes are digested with
restriction enzymes to generate DNA fragments that are then separated by gel
electrophoresis based on their size. In TRFLP, PCR products are generated with
5ʺ-fluorescent-labeled primers before endonuclease digestion to generate the termi-
nal restriction fragments (Marsh, 1999). The mixture of DNA fragments can then
be analyzed in an automated DNA sequencer, but only the terminal fragments with
labels can be detected and displayed. Several bioinformatics methods have been
developed for peak identification and taxonomic assignment for TRFLP results,
such as T-REX (T-R FLP analysis EX pedited) (Culman et al., 2009), web-based
TRFMA (Nakano et al., 2006), and Excel-based tools (Fredriksson et al., 2014).
TRFLP simplifies complex banding pattern analysis compared to PCR-RFLP and
allows for the more rapid identification of microbial composition in a community.
Although RFLP-based methods are able to provide fingerprint patterns to detect
community structure, problems exist, including the introduction of PCR bias and
variation caused by the choice of restriction enzymes (Osborn et al., 2000; v.
Wintzingerode et al., 1997). Phylogenetic oligonucleotide arrays, an amplification-
independent method, were developed to overcome these pitfalls. PhyloChip, devel-
oped at Lawrence Berkeley National Laboratory, is the most comprehensive and
high-density phylogenetic oligonucleotide array to detect the presence of bacteria and
archaea from direct microbial DNA or PCR products with a broad dynamic range.
The platform developed its own proprietary bioinformatics pipelines to identify vari-
ations in 16S rRNA gene sequences. The latest version of PhyloChip (PhyloChip G4),
currently available from Second Genome, Inc., can identify and measure the relative
abundance of more than 50,000 individual microbial taxa. PhyloChip has been used
to characterize microbial communities in a variety of samples and environments. In
one case, spatial patterns of microbial species in a rough environment, salt crusts
8 The Gut Microbiome: Bench to Table
of the hyper-arid Atacama Desert, were examined using PhyloChip G3. The results
from this study identified two dominant species, Halobacteriales and Bacteroidetes,
and the existence of a few algal and cyanobacterial species in low abundance levels
(Finstad et al., 2017) in this extreme habitat.
FISH is another powerful method that evolved from non-FISH in 1980 (Bauman
et al., 1980). FISH targeting microbial SSU rRNA genes can be used to determine the
spatial distribution and genome structure of microbes. Typically, probes targeting the
16S rRNA gene are generally 18–30 nucleotides in length with fluorescence labels.
These probes can penetrate through the cell membrane and eventually hybridize to
the target 16S rRNA genes in situ (Hugenholtz et al., 2002). Hybridized target genes
within the individual cells can then be visualized using a fluorescence or confocal
microcopy, which allow exploring the spatial location of the targeted genes in addi-
tion to taxonomic identifications. With recent technical innovations, FISH has been
combined with several other cutting-edge biological tools to visualize and elucidate
genome structures of microorganisms. A powerful example is the integration of
FISH with Hi-C to reveal the 3D structure of chromosomes (Abbas et al., 2019). This
method was also used to define the chromosome structure of Mycoplasma pneu-
moniae. By using Hi-C with a 10kb resolution to generate 3D models of the station-
ary M. pneumoniae chromosome and then applying FISH to visualize and measure
distances between genomic regions, Trussart et al. (2017) concluded that transcrip-
tional regulation can be affected by chromosome organization.
Illumina Platform
The Illumina platform relies on the “sequencing by synthesis” technology invented
by Solexa (Bentley et al., 2008). In contrast to various costly enzymes used by
pyrosequencing in the 454 platform, sequencing by synthesis uses only DNA poly-
merase. Specifically, in the flow cell, DNA templates are amplified using isother-
mal bridging amplification, which relies on arching over and hybridizing to an
adjacent oligonucleotide anchor by DNA strands. Numerous amplification cycles
make the single-molecule DNA template into a clonally amplified arching cluster
of approximately 1,000 clonal molecules. As each Deoxynucleotide triphosphates
(dNTP) is inserted, a fluorescently marked reversible terminator is recorded and
then cleaved to allow the next base to be integrated. The ultimate result is a base-by-
base sequencing that allows a wide range of applications with accurate sequencing
data (Bentley et al., 2008).
Illumina has since acquired this technology from Solexa and marketed several
systems that differ in terms of sequencing capacities. HiSeq (2 × 150 bp) and MiSeq
(2 × 250 ~ 300 bp) series have long been the dominant high-throughput sequenc-
ers. In recent years, Illumina has launched several new systems, such as NovaSeq
6000. The HiSeq X system is the world’s first to break the $1,000 genome barrier for
human whole genome sequencing. The system contains a set of 10 HiSeq X ultra-
high-throughput instruments. Illumina will discontinue its support for HiSeq 2500
and X series after 2023 and 2024, respectively. NovaSeq 6000 is a relatively new
comprehensive Illumina system that enables cost-effective sequencing. Notably, it is
a scalable platform that allows the loading of individual flow cell lanes with differ-
ent library types to suit a wide range of project sizes or needs with the NovaSeq Xp
workflow. It also comes with single and dual flow cell modes with different flow cell
types to generate several combinations of read length and coverage depth, with read
lengths up to 2 × 250 bp (paired-end).
Illumina MiSeq is another powerful sequencer that enables the generation of
approximately 25 million sequencing reads with up to 2 × 300 bp read lengths. In
November 2013, the US Food and Drug Administration approved the marketing of
the NGS system, Illumina’s MiSeqDx, as a diagnostic device for human genome
sequencing. The marketing approval of the first high-throughput genome sequencer
represents a significant step forward in producing genomic data to aid diagnosis and
eventually improve patient care. MiSeq is suitable for amplicon-based microbiome
studies because of its longer read length. Merging its paired-end reads can result in
10 The Gut Microbiome: Bench to Table
Ion Torrent
While the Ion Torrent platform uses sequencing by synthesis and emulsion PCR
somewhat similarly to Illumina chemistry and 454 pyrosequencing, respectively, its
method of detection is unique. If a nucleotide is incorporated into a strand of DNA,
an H+ ion is released. The charge from H+ ions alters the solution pH value, which
can be detected by a proprietary ion sensor. An Ion Torrent sequencer is essentially
a solid-state pH meter. Because the detection is direct and rapid without the need for
scanners, cameras, or light (and without the need for fluorescence and chemilumines-
cence reagents), the cost of sequencers is low, and running time is short (<7 hours).
While high error rates in sequencing homopolymer stretches and repeats (similar to
the 454 platform) are undesirable, Ion Torrent generates a mean read length up to 400
bp, which is quite appealing for SSU amplicon sequencing.
Salipante et al. (2014) compared the performance of two common benchtop NGS
sequencers, Illumina MiSeq and Ion Torrent Personal Genome Machine (PGM), for
Tools and Resources for Microbe Analysis 11
Given limited read lengths, only one or two adjacent HV regions of the 16S rRNA
gene can be sequenced using the second-generation sequencing technologies. Short
reads may not be effective for species- or subspecies-level identification. New plat-
forms with long read sequencing, also termed the third-generation sequencing tech-
nologies—such as those developed by Pacific Biosciences (PacBio) and Oxford
Nanopore—have increasingly gained popularity.
PacBio Sequel Systems use a single-molecule, real-time sequencing technology
to provide highly accurate long reads. Single-molecule, real-time cells contain mil-
lions of zero-mode waveguides (ZMWs). A single DNA molecule is immobilized in
each ZMW and is repeat sequenced with real-time detection of base incorporation.
Each of the four nucleotides A, C, G, and T are labeled with different fluorescent
dyes on their terminal phosphates. Light is emitted during DNA synthesis through
the template in the ZMW and recorded as nucleotide sequences. The sequencing
reads can reach up to tens of Kb. However, one key issue with the technology in
the past is a high sequencing error rate, sometimes up to 11%–15%. PacBio tries to
reduce the error rate by utilizing a consensus accuracy function to reduce system-
atic errors. Specifically, both DNA strands in one ZMW can be sequenced multiple
times to generate multiple reads (subreads); a consensus sequence of these subreads
can yield higher accuracy (Rhoads and Au, 2015). The newest release is the Sequel
II System, which produces exceptional results with faster running time, affordable
expense, and highly accurate long reads. PacBio has recently been successfully used
in SSU sequencing projects to cover the full length of 16S rRNA genes (Callahan et
al., 2019; Earl et al., 2018).
Oxford Nanopore is another promising DNA sequencing technology designed
to have essentially no reagent costs, little sample preparation, and ultra-long read
length to reduce the need for complicated assembly algorithms and reveal the long-
range structure of the genome at the same time (Branton et al., 2010). The prin-
ciple of Nanopore is that when a strand of DNA passes through a nanopore, the
current is changed as the bases T, C, G, and A pass through in different combina-
tions. Several sequencers, such as MinION, GridION, and PromethION, that differ
in device size, nanopore channels, and sample prep procedures are currently avail-
able. Nanopore technology has also been successfully applied to sequence full-length
16S rRNA genes. One study analyzed the mouse gut microbiome by full-length 16S
rRNA amplicon sequencing using Nanopore and compared the results with short-
read sequencing data (Shin et al., 2016). The study found no significant differences
12 The Gut Microbiome: Bench to Table
in major taxonomic assignment between the two methods. However, the differ-
ence becomes apparent at species-level identification. Empowered with long reads,
Nanopore is able to identify more species than short-read platforms, which improves
the classification accuracy of community bacterial composition (Shin et al., 2016).
It is conceivable that this technology will play a critical role in field microbial ecol-
ogy and microbiome studies with anticipated significant reduction of the sequencing
error rate in the near future.
Greengenes is a database for full-length 16S rRNA genes of bacteria and archaea
that aims to provide a curated taxonomy for users based on de novo tree inference
(DeSantis et al., 2006; McDonald et al., 2012). The latest Greengenes version is
gg_13_8, released in August 2013, and is a minor improvement over the previous
version (the gg_13_5 released in May 2013). The improvement addressed the missing
genus and species names. If a genus or species name is missing for a particular OTU,
the National Center for Biotechnology Information (NCBI) taxonomy of cluster
members was checked, and the most supported names were then selected to update
the taxonomy of representative sequences. The taxonomic name was then transferred
to all the lower similarity representative sequences. The number of sequences in
Greengenes and other databases is listed in Table 1.1.
Although Greengenes has not been updated since 2013, this curated, chimera-
checked database is still robust and is one of the most used databases in 16S gene
sequencing analysis. Several analysis tools, such as the Quantitative Insights into
Microbial Ecology (QIIME) analysis pipeline and the metagenome function prediction
TABLE 1.1
Commonly Used Databases on Marker Gene–Based Microbiome Studies and
Their Number of Sequences
Database Latest Version Release Date Number of Sequences
Greengenes gg_13_8 Aug 2013 1262,986 (16S rRNA sequence)
SILVA SILVA 132 Dec 2017 2,090,668 (SSU Ref), 695,171 (SSU Ref NR)
RDP Release 11.5 Sep 2016 3,356,809 (16S rRNA sequences), 125,525
(28S rRNA sequences)
EzBioCloud Version 20191112 Nov 2019 65,050 (16S rRNA sequences), bacterial genomes
UNITE Version 8.1 Sep 2019 98,183 (fungal SH with DOIs at 1.5% threshold)
PR2 Version 4.12.0 Aug 2019 183,949 (eukaryotic SSU)
Tools and Resources for Microbe Analysis 13
SIL
SILVA (from Latin silva, forest) is a comprehensive rRNA gene web resource dedicated
to providing the taxonomy information for bacteria, archaea, and eukarya (Pruesse
et al., 2007; Quast et al., 2012). It contains the ribosomal RNA sequence datasets of
aligned small (16S/18S, SSU) and large subunit (23S/28S, LSU) rRNA sequences,
which are quality checked and regularly updated. The latest full release is the SILVA
SSU/LSU version 132, released in December 2017. The preliminary release informa-
tion of SILVA 138 is available on its website, and the version is expected to launch
around the end of 2019. SILVA is highly recommended by the widely used analy-
sis platform mothur as its default reference database. The SILVA database contains
different sections of rRNA genes. “SSU Parc” includes all aligned sequences with
an alignment identity value ≥ 50, alignment quality value ≥ 40, and base-pair score
or sequence quality ≥ 30. “SSU Ref” was created by excluding sequences shorter
than 1,200 bases (for bacteria and eukarya) and below 900 bases (for archaea) or an
alignment identity lower than 70 or an alignment quality value lower than 50 from
the SSU Parc dataset. Additionally, SILVA offers a nonredundant (NR) version of
the SSU Ref dataset, “SSU Ref NR,” where highly identical sequences are removed
by applying UCLUST with a 99% identity criterion, yet sequences from cultivated
species have been kept in all cases. By adding all sequences to the SSU Ref tree of
SILVA release 128, a guide tree was calculated. Similarly, for LSU rRNA databases,
SILVA also contains “LSU Parc” and “LSU Ref” datasets based on different criteria
of LSU gene sequences. “LSU Ref” also contains a calculated guide tree.
In addition, SILVA provides several online data processing and analysis functions,
including probe and primer evaluation with “TestProbe” and “TestPrime,” sequence
alignment and classification, and an online data analysis service, SILVAngs, for
rRNA NGS amplicon reads. SILVAngs uses the taxonomies and alignments of the
SILVA rRNA gene database, supports reads classification, and offers many user-
friendly output files in table, chart, and sequence formats.
The ribosome database project (RDP) offers quality-controlled bacterial and archaeal
SSU rRNA sequence alignments and annotation, as well as data analysis tools.
Besides the SSU rRNA genes of bacteria and archaea, the RDP also adds a collection
of fungal LSU 28S rRNA sequences (Cole et al., 2009; Cole et al., 2013). The latest
release is RDP 11.5, which was released in September 2016. The RDP implements
improved alignments and also provides collections of tools for rRNA analysis. Newly
obtained sequences are aligned and classified with the RDP Aligner and Classifier
with its own phylogenetic assessment. Specifically, the RDP utilizes the infernal sec-
ondary structure-aware aligner to improve its alignment quality, speed, and con-
sistency. The RDP Classifier is a powerful and widely used method in taxonomy
14 The Gut Microbiome: Bench to Table
classification for SSU rRNA data analysis. The RDP Classifier and RDP Hierarchy
Browser Bacteria and Archaea hierarchy model has been updated to training set
no. 16, to which over 300 new genera and 2000 new sequences have been added.
Notably, the RDP provides its resource files for the NCBI LinkOut service, which
enables users to switch from the sequence records in the Nucleotide and BioProject
databases of the NCBI directly to the corresponding RDP sequence records. The
RDP also provides an online RDPipeline for high-throughput amplicon analysis and
offers multiple tools, such as primer-probe examination, approximate phylogenetic
building of user-submitted sequences, chimera removal, and automated alignment
(Cole et al., 2013).
UNITE
The ITS region is the most widely used fungal marker to identify fungi and explore
fungal diversity in the environment. The UNITE is a web-based database and
sequence management environment dedicated to the molecular identification of
fungi using ITS regions as markers (Nilsson et al., 2018). UNITE is arguably the
most commonly used database for classifying and annotating fungi with ITS high-
throughput sequencing. It includes ~1000,000 public fungal ITS sequences that
are clustered into ~459,000 species hypotheses for reference and assigned DOIs to
reduce ambiguity among studies. UNITE redesigned how it handles unclassifiable
species hypotheses, integrated the Global Biodiversity Information Facility’s taxo-
nomic backbone, and supports parallel taxonomic classification systems (Nilsson et
al., 2018). Users can also compare sequences to species hypotheses through Basic
Local Alignment Search Tool (BLAST), probabilistic method for taxonomical
Tools and Resources for Microbe Analysis 15
classification (PROTAX) (Abarenkov et al., 2018) and other query tools through
UNITE. In addition to taxonomic annotation, UNITE also adds relevant metadata,
such as the host of collection and voucher specimens. Importantly, UNITE supports
web-based third-party annotations to update taxonomy and nomenclature informa-
tion and to correct suboptimal taxonomic annotation and other metadata across pub-
lic DNA sequences (Nilsson et al., 2018). With these efforts, UNITE identifies fungi
from ITS sequence data with assembling and disseminating taxonomic, ecological,
and geographical metadata. UNITE acts as a data provider for a number of metaba-
rcoding software tools and exchanges data with all major fungal sequence databases
and other community resources on a regular basis.
Pr2
The Protist Ribosomal Reference (PR2) database offers exclusive access to the ribo-
somal RNA and DNA sequences of eukaryotic SSU 18S with curated taxonomy
(Guillou et al., 2013). The majority of sequences in this database are nuclear-encoded
protistan sequences. Yet PR2 also contains sequences from metazoans, land plants,
macrosporic fungi, and eukaryotic organelles (mitochondrion, plastid, and others),
which is useful for high-throughput sequencing data analysis. PR2 also carefully
checks introns and putative chimeric sequences. Additionally, there are web tools
one can use to search the database by sequence similarity (Guillou et al., 2013).
DNA sequencing
Targeted Hypervariable Primer selecon Sequencing plaorm
regions F515/R806, Illumina MiSeq v3
Full length, V3-V4, V4 F341/R805 chemistry, 2x 250 cycles
Pre-processing
Remove primers, demulplex, quality filter
OTU/ASV detecon
QIIME, Mothur, FROGS, iMAP
FIGURE 1.1 Recommended data analysis pipeline for marker gene-based microbiome studies.
Tools and Resources for Microbe Analysis 17
each OTU to represent the whole cluster. The default is to pick the most abundant
sequence from a cluster as its representative, as these sequences are less likely to
represent sequencing errors. When uclust or usearch is selected, the cluster seed is
used as the representative sequence. The QIIME team also recommends the addition
of chimera checking after OTU picking by UCHIME to exclude chimera in down-
stream analyses. The subsequent step in the pipeline includes assigning taxonomy
to representative sequences. The latest Greengenes database is used as the default.
The sequence alignment and phylogeny construction can then be performed with
Python Nearest Alignment Space Termination (PyNAST) and FastTree by default,
respectively. An OTU table is finally generated in the Biological Observation Matrix
(BIOM) format. For downstream analyses, OTU tables and phylogenetic trees gener-
ated from upstream analysis and corresponding mapping files are used. A second-
level quality filtering can be done to reduce the problem of spurious OTUs, followed
by a rarefaction step at a user-defined subsampling depth. After that, the OTU table
is collapsed into taxa to be summarized at each taxonomic level. A series of plots
including bar, pie, and area graphs can be generated and then visualized via a web
interface. QIIME pipeline integrates two diversity analyses, alpha and beta diver-
sity. Three alpha diversity metrics, phylogenetic diversity (PD)_whole_tree, Chao1,
and observed_OTU, are the default outputs. Dozens of other metrics are optional
in alpha_diversity.py script, for example, Simpson and Shannon indices. QIIME
recommends Unifrac (Lozupone and Knight, 2005) for beta diversity analysis and
generates weighted and unweighted Unifrac to analyze community differences.
Additionally, interactive diversity graphs, such as rarefaction curves based on alpha
diversity metrics and principal coordinate analysis (PCoA) based on community beta
diversity distance can be accessed through the output HTML documents. Further
statistical analysis of taxa and diversities can be performed within QIIME or with
external methods. In addition to common t-tests, QIIME also offers multivariate
analyses in the script compare_categories.py, including analysis of similarities
(ANOSIM) and Adonis, to examine beta diversity. OTU network analysis is also
integrated in QIIME.
Since 2017, the QIIME team has reengineered and developed a new system,
QIIME 2 (Bolyen et al., 2019). As a result, the conventional QIIME or QIIME 1 is no
longer officially supported. QIIME 2 also integrates third-party tools in the form of
plugins to allow others to contribute to its function. QIIME 2 provides better support
for paired-sample and time series analyses. Although several features in QIIME 1
are retained, QIIME 2 offers several key improvements over QIIME 1. One critical
aspect of QIIME 2 is using ASV instead of OTU as microbial features. The recom-
mended demultiplexing and denoising workflow in QIIME 2 for AVS identification
is the DADA2 algorithm. Deblur is also used as an optional denoising method to
combine q2-Vsearch to join reads before proceeding. Specifically, these denoising
methods exclude noisy sequences, correct marginal sequence errors (in DADA2),
perform chimera removal and singleton removal, join denounced paired-end reads
(in DADA2), and perform sequence dereplication. Both DADA2 and Deblur contain
internal chimera removal methods and abundance filtering during the denoising pro-
cess. However, QIIME 2 still keeps three OTU-picking options, similar to QIIME 1.
UCHIME and Vectorized search (Vsearch) are used to remove chimera sequences
18 The Gut Microbiome: Bench to Table
mothur
mothur is another widely used data pipeline for amplicon-based microbial commu-
nity analysis. As a comprehensive pipeline, mothur integrates several preexisting
tools and incorporates additional features to process raw community sequencing data
(Schloss et al., 2009).
Supporting documents and a discussion forum that contain procedures to pro-
cess sequence data generated from Sanger, PacBio, Ion Torrent, 454, and Illumina
platforms and provide corresponding standard operating procedures are available
for mothur users and developers. For example, the MiSeq standard operating pro-
cedure contains the procedures for raw sequence demultiplexing, quality control,
chimera removal, taxonomic classification, OTU clustering, diversity analysis, and
phylogeny-based analysis (Kozich et al., 2013). mothur merges forward and reverse
reads to make contigs and removes ambiguous sequences. Unique sequences are
then identified and aligned to the reference. mothur provides several alignment
databases containing 16S and 18S gene sequences compatible with the Greengenes
or SILVA alignments. mothur removes chimeras using the Vsearch algorithm and
identifies nonbacterial lineages, such as those related to chloroplasts and mito-
chondria. Multiple algorithms are available for sequence clustering, including
Opticlust (default), Vsearch distance-based greedy clustering, Vsearch abundance-
based greedy clustering, furthest neighbor, average neighbor, nearest neighbor, and
Unique. The pipeline also performs rarefaction and microbial diversity analyses. A
numbers of diversity metrics, such as ACE, Chao1, observed OTU, Simpson, and
Shannon, can be calculated. Distance metrics, such as weighted complete lineage,
unweighted Unifrac, and Jaccard, are optional with various commands. mothur
also contains tools for ordination analysis using PCoA and nonmetric dimensional
scaling (NMDS). The diversity results can be statistically tested within mothur
using correlation measurements of the relative abundance of each OTU or analy-
sis of molecular variance. Additionally, mothur implements nonparametric analysis
tools—Metastats and linear discriminant analysis (LDA) effect size (LEfSe)—to
examine if there are significantly different OTUs between groups of interest. mothur
workflows are also available in Galaxy.
Both QIIME and mothur share numerous functional features in sequence data
preprocessing, OTU clustering, and diversity index extraction, as well as the genera-
tion of OTU or feature tables. The major difference lies in interactive visualizations.
Tools and Resources for Microbe Analysis 19
FROGS
Find, Rapidly, OTUs with Galaxy Solution (FROGS) is a recently developed Galaxy-
supported pipeline that aims to analyze large sets of amplicon sequences (Escudié
et al., 2017). The pipeline includes a variety of functions, such as preprocessing, cluster-
ing, chimera removal, filtering, and taxonomic or phylogenetic affiliation assignment,
visualization, and tree construction. FROGS handles both single and paired-end
data from the Illumina platform and those generated using 454. The pipeline merges
paired-end reads using Flash with 10% mismatch tolerance and ambiguous reads
removal. Dereplication is also achieved in this step, and a graphical report is gener-
ated for posterior cleaned sequences. The pipeline then uses Swarm for sequence
clustering and Vsearch with de novo UCHIME to remove chimeras. Notably, an
innovative cross-sample validation process is applied to validate the chimeric status
on all samples. In each sample, chimeras are first identified separately, but in the
end, this sequence is considered chimeric only if it is marked as a chimera in all
samples where it exists. This is different from mothur, where only the redundant chi-
mera sequences are removed. FROGS also applies an abundance filter to screen clus-
ters and optionally delete clusters not present in all replications. Taxonomy can be
assigned up to the species level with the implemented RDP Classifier or BLASTn+.
Common SSU marker gene databases, such as Greengenes, SILVA, and EzBioCloud,
can be selected as the reference database. One unique characteristic of FROGS is
that it considers conflicting affiliations with BLASTn+, and if affiliation results find
multiple hits with different taxonomy, the first level of confliction and lower taxono-
mies are denoted as “multi-affiliation” in the final OTU table. Because of its modular
design, which allows users to choose their tools of preference or processing order,
FROGS is a fast, scalable, and parallelizable pipeline (Escudié et al., 2017).
FROGS allows in-depth analyses not only of 16S, 18S, and 23S rRNA genes but
also of amplicons from functional genes, such as dsrB. The latest version of FROGS
can also be used to analyze ITS data. The entire pipeline can be run in either com-
mand line or in Galaxy platform, which is user friendly for those without much bio-
informatic experience. FROGS has the advantage of using Swarm and its adaptive
sequence agglomeration instead of using a global similarity threshold. Additionally,
FROGS has a rigorous step of removing chimera and explicitly considers conflicting
20 The Gut Microbiome: Bench to Table
affiliations. Compared with QIIME, mothur, and UPARSE, FROGS performs better
than mothur and is more conservative than QIIME on the V4 region and better on
V3/V4 regions for in silico datasets. Furthermore, it performs relatively better for
staggered abundances and worse for uniform ones and produces less spurious OTUs
than other pipelines on real datasets (Escudié et al., 2017).
iMAP
The Integrated Microbiome Analysis Pipeline (iMAP) is a very recent development
that provides the research community with a user-friendly and portable tool that inte-
grates bioinformatics analysis and data visualization (Buza et al., 2019). The pipeline
includes bundles of commands wrapped individually in driver scripts for performing
various analyses and data visualization. It requires data files (sample metadata and
raw sequences), software, and reference databases to be in place before execution via
either a command line interface or from a Docker container command line interface.
Outputs from major steps can be conveniently transformed, visualized, and summa-
rized into a progress report. The default reference databases are up-to-date SILVA
seed or Greengenes for mothur and QIIME, respectively.
One of the interesting features of iMAP is that it allows exploratory analysis of
sample metadata or experimental/environmental variables. iMAP uses seqkit for
the general inspection of raw sequences, FastQC for base quality assessment, and
BBDuk for trimming and filtering low-quality or control reads. iMAP uses standard
approaches in mothur (default) and QIIME 2 for sequence processing and classifi-
cation. Moreover, it applies additional quality control and removes all undesirable
matches including the unknown and any sequences classified to nonbacterial lin-
eages, such as archaea, chloroplasts, eukaryotes, mitochondria, and viruses.
iMAP uses a fairly unique approach for OTU clustering and taxonomy assign-
ment. It relies on a combination of phylotype, OTU-based, and phylogeny methods
to assign conserved taxonomy to OTUs. Merged quality sequences are binned into
known phylotypes up to the genus level. All sequences are binned into clusters of
OTUs based on their similarity at ≥97% identity, and precision and the false dis-
covery rate (FDR) are then calculated using Opticlust, a default mothur function for
assigning OTUs. In the phylogeny method, a tree that displays consensus taxonomy
for each node is generated. The output from all three methods—phylotype, OTU-
based, and phylogeny—is manually reviewed, deduplicated, and integrated to form a
complete OTU taxonomy output.
The pipeline uses the R package for microbial diversity analysis. At least three
ordination methods—principal component analysis (PCA), PCoA, and NMDS—are
provided. Furthermore, phylogenetic annotation is done using Interactive Tree Of
Life (iTOL) tree viewer with the aid of selected annotation files, such as the species
richness, diversity, and relative abundances files.
While the iMAP pipeline uses an integrated approach for OTU clustering and
taxonomy assignment and demonstrates improvements over some widely used pipe-
lines, such as mothur and QIIME, it offers limited solutions for downstream analysis
beyond microbial diversity and a phylogenetic tree view, such as the identification
of differential abundant taxa and biomarker discovery using advanced statistical
Tools and Resources for Microbe Analysis 21
groups, they are also used to detect significant taxa, for example, in a study involv-
ing HIV-positive patients with or without antiretroviral therapy and healthy con-
trols (McHardy et al., 2013). Similarly, nonparametric counterparts of these tests,
including the Wilcoxon rank sum test and the Kruskal–Wallis test by rank, are also
widely used in microbiome studies. LEfSe is an algorithm for high-dimensional bio-
marker discovery and identification of genomic features such as genes, pathways, or
taxa under experimental or pathophysiological conditions (Segata et al., 2011). The
algorithm includes three major steps—detecting features (taxa or ASV) significant
in relative abundance using the nonparametric Kruskal–Wallis test, investigating
biological consistency using the Wilcoxon rank sum test, and using LDA to estimate
the effect size of each differentially abundant feature. The method has been widely
used for microbiome data analysis, including the discovery of biomarkers or dif-
ferentially abundant taxa derived from OTU or ASV tables or biological pathway
data inferred using PICRUSt (Kageyama et al., 2019; Lo Presti et al., 2019; Puri et
al., 2018; Sims et al., 2019). The popularity of the method is partially driven by the
implementation of a user-friendly Galaxy version of the algorithm (http://hutten-
hower.sph.harvard.edu/galaxy/).
In several regards, OTU or ASV count data are similar to gene expression data
obtained using microarrays or RNAseq. As a result, many algorithms developed in
the early years of the millennium to detect differentially expressed genes, such as
DESeq2 and EdgeR, can be readily adapted to analyze marker gene survey data
(McMurdie and Holmes, 2014). In 2013, Paulson et al. proposed a novel normal-
ization procedure, cumulative sum scaling normalization, to correct the bias in the
assessment of differential abundance introduced by widely used total-sum normal-
ization (Paulson et al., 2013). A zero-inflated Gaussian (ZIG) distribution mixture
model that accounts for biases in differential abundance testing resulting from under-
sampling of the microbial community was also proposed in that paper. Unlike the
ad-hoc heuristic approach used in LEfSe, ZIG uses linear modeling and has been
shown to be more sensitive under simulated conditions. More differentially abundant
species can be identified using ZIG than LEfSe in real microbiome datasets (Paulson
et al., 2013). However, this method does not address the compositionality issue inher-
ent in marker gene datasets, which poses particular challenges, and traditional statis-
tical tools often become invalid.
In 2014, an algorithm dealing with compositional data typically seen in data-
sets involving multiple omics approaches was developed. This method, ALDEx2
(Fernandes et al., 2014), includes several straightforward steps. First, the counts are
converted to probabilities by Monte Carlo sampling from the Dirichlet distribution
with the addition of a uniform prior, usually 1/2. The resultant Monte Carlo Dirichlet
instance is then normalized using the centered log-ratio (CLR) approach. The value
is the base 2 logarithms of the abundance of the feature in each Dirichlet instance in
each sample divided by the geometric mean abundance of the Dirichlet instance of
the sample. Significance tests between groups of interest are then conducted using the
Welch’s t-test and the Wilcoxon rank sum test; P values obtained are corrected using
the Benjamini–Hochberg procedure; and the effect size is also estimated. Compared
to traditional statistical test tools, such as the widely used LEfSe, ALDEx2 tends
to reduce the number of false positives, especially when the sample size is small
Tools and Resources for Microbe Analysis 23
(Fernandes et al., 2014). Moreover, the method enables a unified analysis for high-
throughput sequencing datasets, including marker gene count tables, RNAseq, and
ChIPseq datasets.
Recently, a novel statistical framework called analysis of composition of micro-
biomes (ANCOM) has been developed to detect taxa differing significantly in two
or more populations or experimental groups (Mandal et al., 2015). The framework
is based on Aitchison log-ratios without distributional assumptions and is therefore
quite flexible. ANCOM uses the W statistic and selects an internal threshold (W val-
ues) for significance (no P values provided). One of the limitations of the framework
is the assumption that less than 25% of the taxa are changing significantly between
two groups, which limits its application when two populations or groups under com-
parison have drastic differences. Nevertheless, when compared to the conventional
t-test and the aforementioned ZIG method, ANCOM can reduce false positives by as
much as 68% or even more while improving detection power. Since its advent, the
method has become rapidly accepted (Iszatt et al., 2019; Peterson et al., 2019) and is
now implemented in the QIIME pipeline.
A detailed comparison of several methods designed for differential abundance
detection (originally intended for gene expression count data obtained using micro-
arrays or RNAseq) under various scenarios, including sample size, effect size, and
distributional assumptions, has been recently conducted (Hawinkel et al., 2017). For
example, the sensitivity of both ANCOM and ALDEx2 is fairly low (<25%) under
the negative binomial distribution with or without correlation. Of the methods tested,
DESeq2 and SAMseq have modest sensitivity when the sample size is small, while
metagenomeSeq is more powerful with a sensitivity > 50% when the sample size is
larger than 25. However, all methods fail to control the FDR. Compared to ALDEx2,
ANCOM has a much higher FDR under all the scenarios tested, which is in sharp
contrast to the original claim that the method has reasonable power to detect differ-
entially abundance log-ratios while controlling the FDR at a nominal level (<0.05).
Other studies have also suggested that ANCOM is unable to control the FDR, espe-
cially under the global null setting (Brill et al., 2019). Moreover, ANCOM is com-
putationally intensive when used at the OTU level without filtering out very rare
features. Weiss et al. (2017) compared several methods along with ANCOM and sug-
gested that ANCOM behaves better in terms of sensitivity and controlling the FDR
than those methods not specifically designed for handling compositionality.
During the preparation of this chapter, we conducted a direct comparison of sev-
eral popular methods using a published 16S rRNA gene dataset (Liu et al., 2019)
(Sequence Read Archive (SRA) accession: PRJNA534501). The part of the dataset
used for this comparison included two experimental groups, normal control mice
(N = 10) and colitis mice induced with dextran sulfate sodium (DSS) (N = 10). The
sample OTU matrix includes 20 × 1,666 data points, a typical dataset commonly seen
in controlled animal experiments.
The number of significant genera identified by all four non-ANCOM–related
methods is 22, whereas the number of significant genera identified by all six methods
is 15 (Table 1.2); 15 of the 16 genera identified by ANCOM (v2) are also detected by
all other methods. ALDEx2 performs well compared to other methods. For composi-
tional datasets, such as those obtained in marker gene–based microbiome studies, we
24 The Gut Microbiome: Bench to Table
TABLE 1.2
The Number of Differentially Abundant Genera Identified by
Six Algorithms Tested
ANCOM_ ANCOM_
Version Method LEfSe v1 v2 Wilcoxon DESeq2 ALDEx2
v1.0 LEfSe 35 23 16 31 24 24
v1.1-3 ANCOM_v1 23 16 22 21 20
v2.0 ANCOM_v2 16 15 15 15
v1.0 Wilcoxon 31 24 24
v1.12.4 DESeq2 25 22
v1.4.0 ALDEx2 24
All algorithm except LEfSe was at a cutoff value of the FDR < 0.05. The default cutoff for LEfSe was used
(absolute LDA > 2.0). The values in off-diagonal entries indicate the number of significant genera identi-
fied by both methods.
would recommend ALDEx2 as a default option for detecting taxa with significantly
different abundance between groups of interest.
This software relies on meta-analytic principles and provides tools to use the results
of OTU-level significance tests (with meaningful one-sided P values) to identify and
visualize branches in a phylogenetic tree that are significantly responsive to experi-
mental interventions or changes in environmental conditions. SigTree uses P values
rather than raw data and allows users to select models appropriate for specific study
designs, including accounting for study-specific data distribution (Poisson, negative
binomial, or nonparametric). The package also provides two multiple testing cor-
rection options, the Hommel and Benjamini–Yekutieli corrections. Together, the
authors claim these flexible features allow SigTree to be applicable in any experi-
mental design and with any high-throughput technology. Compared to Gneiss, which
does not include convenient tree-level visualization tools, SigTree provides better
tree visualization. Moreover, SigTree does not have the constraints typically associ-
ated with the isometric log-ratio transformation used in Gneiss, including issues with
zero replacement.
A novel approach superficially similar to Gneiss has been proposed to identify
balances or microbial signatures, a group of taxa that can better predict treatment out-
comes or a phenotype of interest (Rivera-Pinto et al., 2018). This package, selbal, uses
a greedy stepwise algorithm for the selection of balances that preserves the principles
of compositional data analysis. The process of balance identification by selbal includes
two steps, modeling the response variable and identifying the smallest number of
taxa with the highest prediction or classification accuracy. The software also provides
several choices for zero replacement, including the default geometric Bayesian mul-
tiplicative replacement method. The variables can be categorical (dichotomous) or
numerical (continuous). To demonstrate its utilities, we analyzed the aforementioned
dataset (Liu et al., 2019) (SRA accession: PRJNA534501) using selbal with default
parameters. Three variables are considered, treatment (dichotomous, normal, or DSS),
spleen weight (g, numerical), and colon length (cm, numerical). The selbal algorithm
selects 20 variables (genera) with good classification accuracy that are compatible
with the 24 genera identified using ALDEx2. As Figure 1.2a shows, a microbial signa-
ture consisting of the balance (log ratio) of Streptococcus (numerator) to an unclassi-
fied genus in the family Peptostreptococcaceae (denominator) can readily distinguish
between DSS-induced colitis mice and normal control mice. The balance value for
DSS mice is negative, suggesting that the relative abundance of the unclassified genus
in Peptostreptococcaceae is much higher than that of Streptococcus. The cross-vali-
dation data indicate that this global balance is very robust and is selected 62% of the
time during the validation. Moreover, a balance, consisting of four genera, an unclas-
sified genus in the family Clostridiaceae and the genus Clostridium (numerator) and
Streptococcus and [Clostridium] in the family Peptostreptococcaceae, has a strong
predictive power for the spleen weight (R2 = 0.818) with a very low mean squared error
of 0.0041 (Figure 1.2b). Two of the four genera, Clostridium and Streptococcus, are
included in balance selection more than 60% of the time, further indicating that the
selected global balance is robust. In addition, the algorithm also identifies a microbial
signature for the colon length, but the association is modest with R2 = 0.47.
26 The Gut Microbiome: Bench to Table
FIGURE 1.2A A microbial signature consisting of the balance (log ratio) of Streptococcus
(numerator) to an unclassified genus in the family Peptostreptococcaceae (denominator).
FIGURE 1.2B Regression Analysis of the microbial signature in association with spleen weight.
interaction network inference based on network theory and practice provides system-
level insight into potential ecological relationships of microorganisms in a microbial
community. For example, modularity, the extent to which microbial interactions are
organized into modules or subnetworks in a global network, may reflect habitat het-
erogeneity or phylogenetic clustering of closely related microbes, and modules with
closed-linked species may represent some key coevolution units (Olesen et al., 2007).
Furthermore, knowledge of interaction networks forms the foundation to predic-
tively model the interplay between environmental factors and microbial populations
(Kurtz et al., 2015) and may hold keys to the development of efficacious probiotics
and antibiotic therapeutics. Ultimately, the knowledge obtained by modeling micro-
bial interactions should facilitate microbial community engineering via the targeted
elimination and expansion of keystone species in a network. Motivated by these lofty
goals, dozens of algorithms have been developed to infer microbial co-occurrence
patterns and model microbial interaction network dynamics (Barberán et al., 2012;
Faust et al., 2012; Freilich et al., 2010; Ruan et al., 2006). For example, a meta-net-
work framework has recently been proposed to decipher microbial interactions and
construct more meaningful networks with biologically important clusters and hubs
(Yang et al., 2019). The framework starts with a loose definition strategy to recover
more correlations before correlation calculation. While direct relationships are dis-
covered, association rule mining is subsequently applied to detect complex forms
of correlations, including indirect correlations using Functional Similarity Weight
(FS-weight) and nonlinear correlations using the part mutual information adjusted by
path consistency algorithm, which enables the detection of interactions that are often
missed using they Pearson and Spearman approaches.
Habitat filtering, the co-occurrence of microbes due to habitats rather than biologi-
cal interactions being sampled, can induce apparent correlations, resulting in a network
dominated by habitat effects and the masking of true correlations of biological inter-
est (Berry and Widder, 2014). The phenomenon can confound microbial interaction
network inference when samples from different habitats are combined. To overcome
this issue, a novel algorithm to correct for habitat filtering effects has been proposed
(Brisson et al., 2019). This algorithm significantly improves correlation detection accu-
racy compared to Spearman and Pearson correlations, thus enabling the construction
of consensus correlation networks from datasets combining multiple habitats.
Weiss et al. (2016) conducted a comprehensive benchmarking comparison analy-
sis of the performance of eight microbial network inference methods. Commonly
used network inference tools are based on a broad range of underlying methodolo-
gies, such as correlation and regression-based tools (Compositionality Corrected by
REnormalizaion and PErmutation (CCREPE), Sparse Correlations for Compositional
data (SparCC), Random Matrix Theory (RMT)), graphical model inference tools
(Sparse Inverse Covariance Estimation for Ecological Association Inference
[SPIEC-EASI]), extended local similarity analysis (eLSA), and mutual information-
based tools (Mutual Information Clustering (MIC)). Weiss et al. (2016) concluded
that while different tools have various strengths and weaknesses in response to the
diverse challenges presented by microbiome data, all tools result in a high false-
positive rate. An ensemble approach, including CoNet, SparCC, and Spearman and
Pearson methods, tends to enhance the precision of detection. While paired with
28 The Gut Microbiome: Bench to Table
by chance alone and the machine learning–based is the method (Lim et al., 2016) to
extract the interaction information. MPLasso outperforms all methods tested, includ-
ing the aforementioned SPIEC-EASI, SparCC, and CCREPE, as well as Correlation
Inference for Compositional Data through Lasso (CCLasso) and regularized estima-
tion of the basis covariance based on compositional data (REBACCA) in terms of
area under the precision-recall curve and the accuracy (ACC) of network association
prediction in synthetic datasets. In the presence of prior knowledge, MPLasso can
achieve remarkably high accuracy in terms of the edge recovery rate, up to 95%.
Moreover, MPLasso can robustly and accurately estimate microbial interactions,
with reproducibility up to 90% in real microbiome datasets.
Microbial communities are dynamic in nature. The abundance of community
members fluctuates temporally or in response to environmental factors or perturba-
tions. Similarity-based network inferring tools (Bray–Curtis, Pearson, Spearman, or
LSA) treat time series data as a static snapshot and ignore their temporal dependen-
cies (Alshawaqfeh et al., 2017) and are generally unable to infer nonlinear relation-
ships, especially those with multispecies complex interactions. The Lotka–Volterra
equations, a pair of first-order nonlinear differential equations, have been frequently
used to describe the dynamics of biological systems in traditional macroecology.
Recently, the role of dynamic systems theory in understanding microbial commu-
nity dynamics, especially the utility of generalized Lotka–Volterra models, has been
extensively discussed (Gao et al., 2018; Gonze et al., 2018). For example, using gen-
eralized Lotka–Volterra dynamics modeling, Berry and Widder (2014) found that co-
occurrence networks can recapitulate interaction networks under certain conditions,
but they lose interpretability when the effects of habitat filtering become significant
(Berry and Widder, 2014). In particular, networks suffer from local hot spots of spu-
rious correlation in the neighborhood of keystone or hub species that engage in many
interactions. Recently, a new approach called Learning Interactions from Microbial
Time Series (LIMITS) has been developed (Fisher and Mehta, 2014). LIMITS uses
sparse linear regression with bootstrap aggregation to infer a discrete-time Lotka–
Volterra model for microbial dynamics and overcome some of the obstacles com-
monly encountered while inferring microbial interactions. The obstacles include
the observations that a correlation in the abundance between two microbial species
does not necessarily imply their actual interaction and that it is difficult to infer
parameters in time series data due to the sum constraint on the relative abundance
(compositionality) in typical microbiome datasets. When applied to real microbiome
datasets, LIMITS is able to successfully detect interaction networks dominated by
distinct keystone species.
Alshawaqfeh et al. (2017) proposed a stochastic generalized Lotka–Volterra
dynamic model that adopts the extended Kalman filter (SgLV-EKF) algorithm to
model the microbial interaction network dynamics (Alshawaqfeh et al., 2017). The
proposed stochastic model accounts for the uncertainty in the model by adding a
noise term in the dynamic equation, while extended Kalman filter, due to its ability
to estimate the parameters of nonlinear interactions from a limited number of obser-
vations, is used for the first time to estimate the abundance level of microbes and
their interactions. SgLV-EKF is also compared to five other algorithms, including
two similarity-based (Pearson correlation coefficient and LSA), one integral-based
30 The Gut Microbiome: Bench to Table
(Nelder and Mead, 1965), and two regression-based (Stein’s and LIMITS) algorithms
using both synthetic and real datasets. SgLV-EKF outperforms the other five algo-
rithms in identifying the structure of the interaction network. Moreover, SgLV-EKF
provides robust and reliable performance against the uncertainty in the dynamic
model at an accuracy level higher than 75% and that is also higher than that of
LIMITS, which provides reliable results with consistent accuracy of approximately
60%. Nelder’s algorithm generates results similar to those of SgLV-EKF under vary-
ing measurement noise levels but fails to compensate for randomness in the dynamic
system and is computationally intensive. Stein’s algorithm fails to detect the majority
of the interactions and exhibits very low sensitivity, while the two similarity-based
algorithms show a significant reduction in their accuracy due to the increase in the
process noise power. Moreover, the latter two algorithms lack mathematical model-
ing of the microbial community and are unable to predict network dynamics.
comparison and classification, and functional prediction. The tool suite offers mul-
tiple choices for data scaling and normalization, including CLR transformation. For
visualization, the module provides standard methods for stacked bar or area plots,
interactive pie charts, rarefaction curves, and a phylogenetic tree view. Moreover,
the module provides a heat tree analysis based on the MetaCoder algorithm (Foster
et al., 2017). This unique feature uses the hierarchical structure of taxonomic clas-
sifications to quantitatively and statistically depict taxonomic differences between
microbial communities of interest. Under the community profiling, the suite enables
the generation of six alpha diversity indices—Abundance-based (ACE), Fisher’s
Alpha, Chao1, observed taxa, Shannon, and Simpson. For beta diversity evaluation,
the suite allows two popular ordination methods, PCoA and NMDS, based on five
distance matrixes, Bray–Curtis dissimilarity, Jensen–Shannon Divergence, Jaccard
Index, unweighted Unifrac measures, and weighted Unifrac measures. Moreover,
three algorithms, Permutational multivariate analysis of variance (PERMANOVA),
ANOSIM, and distance-based tests for homogeneity of multivariate dispersions
(Permutational analysis of multivariate dispersions (PERMDISP)), are provided for
global significance testing.
Under the comparative analysis category, multiple algorithms are provided for
users to identify taxa that are significantly different between groups of interest and
to discover potential biomarkers. In addition to classical univariate analysis (such
as t-tests and ANOVA) and nonparametric tests (such as the Mann–Whitney U test
or the Kruskal–Wallis rank sum test), MDP also offers metagenomeSeq, edegR,
DESeq2, LEfSe, and Random Forest.
The MDP module also provides two useful tools that enable functional prediction
from 16S rRNA count tables, PICRUSt for the Greengene database, and Tax4Fun for
the SILVA database. The result is a table containing relative abundance levels of Kyoto
Encyclopedia of Genes and Genomes Orthology. The Kyoto Encyclopedia of Genes
and Genomes Orthology profiles obtained from predictions can be used for functional
diversity profiling based on Kyoto Encyclopedia of Genes and Genomes pathway or
Clusters of Orthologous Groups annotation systems. Because one Kyoto Encyclopedia
of Genes and Genomes Orthology or Clusters of Orthologous Groups can be assigned
to multiple functional groups, MicrobiomeAnalyst offers different approaches to deal
with this issue, including simple sum, normalized sum, and weighted sum methods.
The results are presented as a stacked area plot organized by experimental factors to
help visualize patterns of variations across different conditions. The underlying abun-
dance table is available for downloading. A nice feature of the MicrobiomeAnalyst
suite is that R command line history is shown along with the data in the browser.
One of the unique features of the MicrobiomeAnalyst suite is that it contains a
module for meta-analysis. The Projection with Public Data module allows users to
visually explore their datasets within the context of a compatible public dataset for
context reference and pattern discovery using an interactive 3D PCoA plot. While
there is still room for improvement, the MicrobiomeAnalyst suite is a very user-
friendly online toolbox that enables comprehensive visualization and statistical test-
ing for both marker gene– and whole genome shotgun–based microbiome datasets.
Calypso is a recently developed web application with a powerful yet user-friendly
tool suite for the comprehensive analysis of bacterial, archaeal, viral, and eukaryotic
32 The Gut Microbiome: Bench to Table
communities (Zakrzewski et al., 2017). The package uses count data, metadata, and
an optional matrix of pair-wise community distances (such as Unifrac) as input files
to mine and visualize microbiome environment interactions. The suite emphasizes
multivariate analysis for hypothesis testing, which is one of its key strengths.
The package supports various formats of count data, either a .txt, .csv, or BIOM
file, with taxonomic assignments from phylum to OTU levels. Output files from sev-
eral popular pipelines, such as QIIME (Caporaso et al., 2010), mothur (Schloss et al.,
2009), MG-RAST (Meyer et al., 2008), and MetaPhlAn (Segata et al., 2012), can be
directly uploaded. The metadata file consists of a simple text file in either .txt or.csv
format and contains meta information for each sample, such as sample unique iden-
tifiers, sample groups, sample include/exclude options, and optional explanatory or
environmental variables, such as control vs. treated, disease status, and physiological
parameters. Both numeric and categorical variables are supported.
Calypso provides various tools for data filtering, rarefying, scaling, and transfor-
mation, such as asinh, cumulative sum scaling, log, quantile normalization, square
root, and total sum of squares (TSS). It also recommends the default choice of either
TSS combined with square root transformation or cumulative sum scaling + log2
transformation. Of note, Calypso also provides a CLR transformation, which is one
of the most widely used transformations designed for compositional data.
Calypso provides many graphical tools for data visualization, including bubble
plot, heatmap, interactive hierarchical tree, Krona plot, and standard bar charts, box
plots, and strip charts. Users can exclude samples, select colors, and change figure
resolution and dimensions. Output files include data tables in either .csv or .txt for-
mats and publication-quality images in PNG, PDF, or SVG formats.
Calypso allows users to calculate up to eight alpha diversity indices, such as ACE,
Chao1, Fisher’s Alpha, Evenness, Richness, Shannon, and Simpson. These alpha
diversity measures can be viewed using various bar charts, strip charts, box plots,
scatter plots, dot plots, and interactive boxplots. The significance can be tested using
ANOVA or nonparametric Wilcoxon and Kruskal–Wallis rank sum tests. Complex
associations between microbial diversity and multiple explanatory variables can be
identified by multiple linear regression. Calypso also supports diversity analysis using
mcpHill (Pallmann et al., 2012), which simultaneously investigates several diversity
measures by unifying them in the same mathematical family of indices. The advan-
tage of mcpHill is that it may provide biological insights that would remain hidden
when only examining one arbitrary or a priori committed measure of diversity.
Calypso emphasizes multivariate analysis and provides a broad range of ordi-
nation methods for beta diversity, including unsupervised, such as PCA, PCoA,
detrended correspondence analysis, and NMDS, and supervised methods, such as
redundancy analysis and canonical correspondence analysis, using several distance
matrixes. Moreover, the package offers several methods of global significance test-
ing. For example, associations between microbial community composition and a
single environmental variable can be identified by PERMDISP2 or by comparing
intragroup and intergroup community distances using ANOSIM. More complex
environment–microbiome associations can be identified and tested using redundancy
analysis, correspondence analysis, and PERMANOVA (or Adonis). These methods
Tools and Resources for Microbe Analysis 33
TABLE 1.3
Summarization of Feature Categories of Three Major Web-Based Tools for
Marker Gene Studies.
Category METAGEN-Assist MicrobiomeAnalyst Calypso
Web-based Yes Yes Yes
Implementation
Version v8.84
URL http://www. https://www. http://cgenome.net/calypso/
metagenassist.ca microbiomeanalyst.ca/
Data input OTU table, OTU/ASV table, OTU/ASV table, BIOM file
BIOM file BIOM file
Data processing Scaling, Rarefying, scaling, Rarefying, scaling,
transformation transformation transformation
Quantitative Bar chart, pie Bar chart, pie chart, Bar chart, bubble plot, heatmap,
visualization chart phylogenetic tree, heat scatterplot, strichart
tree
Clustering Dendrogram, Dendrogram, heatmaps Dendrogram, heatmaps, SOM
heatmaps, SOM
Hierarchical tree No Yes Yes
Alpha diversity No 6 indices 8 indices
Beta diversity
Distance matrix Yes Yes Yes
Ordination/ PCA, PSL-DA PCoA, NMDS PCA, PCoA, PLS, RDA, CCA,
multivariate DCA, NMDS
analysis
Global significance No ANOSIM, ANOSIM, PERMANOVA,
testing PERMANOVA, PERMDISP
PERMDISP
Univaraite analysis T-test, ANOVA, T-test, ANOVA, T-test, ANOVA, Wilcoxon,
Wilcoxon, Wilcoxon, Kruskal– Kruskal–Wallis, DESeq2,
Kruskal–Wallis Wallis, edgeR, DESeq2, ANCOM, ALDEx2
MicrobiomeSeq
Feature/biomarker Random Forest, LEfSe, Random Forest LEfSe, Random Forest, LASSO,
discovery SVM stepwise regression, SVM
Functional No PICRUSt and Tax4Fun PICRUSt
prediction
Correlation analysis Pearson, Pearson, Spearman, Pearson, Spearman, Kendall,
Spearman, Kendall, SparCC WGCNA, network analysis,
Kendall factor analysis
Multiple testing No Yes Yes
correction
Regression No Yes Yes
RDA: redundancy analysis; DCA: detrended correspondence analysis; CCA: canonical correspondence
analysis; WGCNA: Weighted correlation network analysis; PLS: Partial least squares; SVM: Support
Vector Machines.
Tools and Resources for Microbe Analysis 35
CONCLUSIONS
The advent of high-throughput sequencing technologies and the rapid development of
user-friendly bioinformatic algorithms have enabled a renaissance of marker gene–
based studies. Freely accessible reference databases have rapidly expanded. Several
user-friendly pipelines, such as QIIME, mothur, and FROGS, have been developed.
Numerous algorithms and sophisticated statistical testing, including machine learn-
ing, have been proposed to detect differentially abundant taxa, identify biomarkers
and taxa correlated with environmental variables, and infer microbial interaction
networks. Easy-to-use web-based tool suites for data analysis have also become
available. However, microbial network inference is still in its infancy, and biological
interpretation and validation of inferred microbial interactions are still extremely dif-
ficult. There are no public databases available for microbial interactions. While many
challenges remain, we strongly believe that knowledge of microbial association and
co-occurrence patterns will establish a foundation to predictively model the interplay
between environmental factors and microbial populations (Kurtz et al., 2015). This
knowledge will ultimately facilitate the development of microbial community engi-
neering by targeted elimination and/or expansion of keystone species in a microbial
community and hold keys to efficacious synbiotic and antibiotic therapeutics.
ACKNOWLEDGMENTS
Mention of trade names or commercial products in this publication is solely for
the purpose of providing specific information and does not imply recommenda-
tion or endorsement by the US Department of Agriculture. The US Department of
Agriculture is an equal opportunity provider and employer.
REFERENCES
Abarenkov, K., Somervuo, P., Nilsson, R.H., Kirk, P.M., Huotari, T., Abrego, N., Ovaskainen,
O., 2018. Protax-fungi: a web-based tool for probabilistic taxonomic placement of fun-
gal internal transcribed spacer sequences. New Phytolologist 220, 517–525.
Abbas, A., He, X., Niu, J., Zhou, B., Zhu, G., Ma, T., Song, J., Gao, J., Zhang, M.Q., Zeng, J.,
2019. Integrating Hi-C and FISH data for modeling of the 3D organization of chromo-
somes. Nature Communications 10, 2049.
Aitchison, J., 1982. The Statistical Analysis of Compositional Data. Journal of the Royal
Statistical Society Series B (Methodological) 44, 139–177.
Alshawaqfeh, M., Serpedin, E., Younes, A.B., 2017. Inferring microbial interaction networks
from metagenomic data using SgLV-EKF algorithm. BMC Genomics 18, 228–228.
Ambardar, S., Gupta, R., Trakroo, D., Lal, R., Vakhlu, J., 2016. High throughput sequencing:
an overview of sequencing chemistry. Indian Journal of Microbiology 56, 394–404.
Arndt, D., Xia, J., Liu, Y., Zhou, Y., Guo, A.C., Cruz, J.A., Sinelnikov, I., Budwill, K., Nesbo,
C.L., Wishart, D.S., 2012. METAGENassist: a comprehensive web server for compara-
tive metagenomics. Nucleic Acids Research 40, W88–95.
Baker, P.I., Love, D.R., Ferguson, L.R., 2009. Role of gut microbiota in Crohn’s disease.
Expert Review of Gastroenterology & Hepatology 3, 535–546.
Balvočiūtė, M., Huson, D.H., 2017. SILVA, RDP, Greengenes, NCBI and OTT — how do
these taxonomies compare? BMC Genomics 18, 114.
36 The Gut Microbiome: Bench to Table
Barberán, A., Bates, S.T., Casamayor, E.O., Fierer, N., 2012. Using network analysis to explore
co-occurrence patterns in soil microbial communities. ISME Journal 6, 343–351.
Bauman, J.G.J., Wiegant, J., Borst, P., van Duijn, P., 1980. A new method for fluorescence
microscopical localization of specific DNA sequences by in situ hybridization of
fluorochrome-labelled RNA. Experimental Cell Research 128, 485–490.
Bentley, D.R., Balasubramanian, S., Swerdlow, H.P., Smith, G.P., Milton, J., Brown, C.G.,
Hall, K.P., Evers, D.J., Barnes, C.L., Bignell, H.R., 2008. Accurate whole human
genome sequencing using reversible terminator chemistry. Nature 456, 53.
Berry, D., Widder, S., 2014. Deciphering microbial interactions and detecting keystone
species with co-occurrence networks. Frontiers in Microbiology 5, 219.
Besser, J., Carleton, H.A., Gerner-Smidt, P., Lindsey, R.L., Trees, E., 2018. Next-generation
sequencing technologies and their application to the study and control of bacterial
infections. Clinical Microbiology and Infection 24, 335–341.
Bolyen, E., Rideout, J.R., Dillon, M.R., Bokulich, N.A., Abnet, C.C., Al-Ghalith, G.A.,
Alexander, H., Alm, E.J., Arumugam, M., Asnicar, F., Bai, Y., Bisanz, J.E., Bittinger, K.,
Brejnrod, A., Brislawn, C.J., Brown, C.T., Callahan, B.J., Caraballo-Rodríguez, A.M.,
Chase, J., Cope, E.K., Da Silva, R., Diener, C., Dorrestein, P.C., Douglas, G.M.,
Durall, D.M., Duvallet, C., Edwardson, C.F., Ernst, M., Estaki, M., Fouquier, J.,
Gauglitz, J.M., Gibbons, S.M., Gibson, D.L., Gonzalez, A., Gorlick, K., Guo, J.,
Hillmann, B., Holmes, S., Holste, H., Huttenhower, C., Huttley, G.A., Janssen, S.,
Jarmusch, A.K., Jiang, L., Kaehler, B.D., Kang, K.B., Keefe, C.R., Keim, P., Kelley, S.T.,
Knights, D., Koester, I., Kosciolek, T., Kreps, J., Langille, M.G.I., Lee, J., Ley, R.,
Liu, Y.-X., Loftfield, E., Lozupone, C., Maher, M., Marotz, C., Martin, B.D., McDonald, D.,
McIver, L.J., Melnik, A.V., Metcalf, J.L., Morgan, S.C., Morton, J.T., Naimey, A.T.,
Navas-Molina, J.A., Nothias, L.F., Orchanian, S.B., Pearson, T., Peoples, S.L., Petras, D.,
Preuss, M.L., Pruesse, E., Rasmussen, L.B., Rivers, A., Robeson, M.S., Rosenthal, P.,
Segata, N., Shaffer, M., Shiffer, A., Sinha, R., Song, S.J., Spear, J.R., Swafford, A.D.,
Thompson, L.R., Torres, P.J., Trinh, P., Tripathi, A., Turnbaugh, P.J., Ul-Hasan, S.,
van der Hooft, J.J.J., Vargas, F., Vázquez-Baeza, Y., Vogtmann, E., von Hippel, M.,
Walters, W., Wan, Y., Wang, M., Warren, J., Weber, K.C., Williamson, C.H.D.,
Willis, A.D., Xu, Z.Z., Zaneveld, J.R., Zhang, Y., Zhu, Q., Knight, R., Caporaso, J.G.,
2019. Reproducible, interactive, scalable and extensible microbiome data science using
QIIME 2. Nature Biotechnology 37, 852–857.
Branton, D., Deamer, D.W., Marziali, A., Bayley, H., Benner, S.A., Butler, T., Di Ventra, M.,
Garaj, S., Hibbs, A., Huang, X., Jovanovich, S.B., Krstic, P.S., Lindsay, S., Ling, X.S.,
Mastrangelo, C.H., Meller, A., Oliver, J.S., Pershin, Y.V., Ramsey, J.M., Riehn, R.,
Soni, G.V., Tabard-Cossa, V., Wanunu, M., Wiggin, M., and Schloss, J.A., 2008. The
potential and challenges of nanopore sequencing. Nature Biotechnology 26, 1146–1153.
Brill, B., Amir, A., Heller, R., 2019. Testing for differential abundance in compositional
counts data, with application to microbiome studies. arXiv preprint arXiv:1904.08937.
Brisson, V., Schmidt, J., Northen, T.R., Vogel, J.P., Gaudin, A., 2019. A new method to cor-
rect for habitat filtering in microbial correlation networks. Frontiers in Microbiology
10, 585.
Buza, T.M., Tonui, T., Stomeo, F., Tiambo, C., Katani, R., Schilling, M., Lyimo, B., Gwakisa,
P., Cattadori, I.M., Buza, J., 2019. iMAP: an integrated bioinformatics and visualization
pipeline for microbiome data analysis. BMC Bioinformatics 20, 1–18.
Callahan, B.J., Wong, J., Heiner, C., Oh, S., Theriot, C.M., Gulati, A.S., McGill, S.K.,
Dougherty, M.K., 2019. High-throughput amplicon sequencing of the full-length 16S
rRNA gene with single-nucleotide resolution. BioRxiv, 392332.
Cani, P.D., Delzenne, N.M., 2011. The gut microbiome as therapeutic target. Pharmacology
& Therapeutics 130, 202–212.
Tools and Resources for Microbe Analysis 37
Caporaso, J.G., Kuczynski, J., Stombaugh, J., Bittinger, K., Bushman, F.D., Costello, E.K.,
Fierer, N., Pena, A.G., Goodrich, J.K., Gordon, J.I., 2010. QIIME allows analysis of
high-throughput community sequencing data. Nature Methods 7, 335.
Case, R. J., Boucher, Y., Dahllöf, I., Holmström, C., Doolittle, W. F., & Kjelleberg, S., 2007.
Use of 16S rRNA and rpoB genes as molecular markers for microbial ecology studies.
Applied and Environmental Microbiology 73, 278–288.
Chakravorty, S., Helb, D., Burday, M., Connell, N., Alland, D., 2007. A detailed analysis of
16S ribosomal RNA gene segments for the diagnosis of pathogenic bacteria. Journal of
Microbiological Methods 69, 330–339.
Cole, J.R., Wang, Q., Cardenas, E., Fish, J., Chai, B., Farris, R.J., Kulam-Syed-Mohideen,
A.S., McGarrell, D.M., Marsh, T., Garrity, G.M., Tiedje, J.M., 2009. The Ribosomal
Database Project: improved alignments and new tools for rRNA analysis. Nucleic Acids
Research 37, D141–D145.
Cole, J.R., Wang, Q., Fish, J.A., Chai, B., McGarrell, D.M., Sun, Y., Brown, C.T., Porras-
Alfaro, A., Kuske, C.R., Tiedje, J.M., 2013. Ribosomal Database Project: data and tools
for high throughput rRNA analysis. Nucleic Acids Research 42, D633–D642.
Crocetti, G.R., Hugenholtz, P., Bond, P.L., Schuler, A., Keller, J., Jenkins, D., Blackall, L.L.,
2000. Identification of polyphosphate-accumulating organisms and design of 16S
rRNA-directed probes for their detection and quantitation. Applied and Environmental
Microbiology 66, 1175–1182.
Culman, S.W., Bukowski, R., Gauch, H.G., Cadillo-Quiroz, H., Buckley, D.H., 2009. T-REX:
software for the processing and analysis of T-RFLP data. BMC Bioinformatics 10, 171.
D’Amore, R., Ijaz, U.Z., Schirmer, M., Kenny, J.G., Gregory, R., Darby, A.C., Shakya, M.,
Podar, M., Quince, C., Hall, N., 2016. A comprehensive benchmarking study of pro-
tocols and sequencing platforms for 16S rRNA community profiling. BMC Genomics
17, 55.
DeSantis, T.Z., Brodie, E.L., Moberg, J.P., Zubieta, I.X., Piceno, Y.M., Andersen, G.L., 2007.
High-density universal 16S rRNA microarray analysis reveals broader diversity than
typical clone library when sampling the environment. Microbial Ecology 53, 371–383.
DeSantis, T.Z., Hugenholtz, P., Larsen, N., Rojas, M., Brodie, E.L., Keller, K., Huber, T.,
Dalevi, D., Hu, P., Andersen, G.L., 2006. Greengenes, a chimera-checked 16S rRNA
gene database and workbench compatible with ARB. Applied and Environmental
Microbiology 72, 5069–5072.
Deshpande, N.P., Riordan, S.M., Castaño-Rodríguez, N., Wilkins, M.R., Kaakoush, N.O.,
2018. Signatures within the esophageal microbiome are associated with host genetics,
age, and disease. Microbiome 6, 227.
Dhariwal, A., Chong, J., Habib, S., King, I.L., Agellon, L.B., Xia, J., 2017. MicrobiomeAnalyst:
a web-based tool for comprehensive statistical, visual and meta-analysis of microbiome
data. Nucleic Acids Research 45, W180–w188.
Duerkop, B.A., Vaishnava, S., Hooper, L.V., 2009. Immune responses to the microbiota at the
intestinal mucosal surface. Immunity 31, 368–376.
Earl, J.P., Adappa, N.D., Krol, J., Bhat, A.S., Balashov, S., Ehrlich, R.L., Palmer, J.N.,
Workman, A.D., Blasetti, M., Sen, B., 2018. Species-level bacterial community profil-
ing of the healthy sinonasal microbiome using Pacific Biosciences sequencing of full-
length 16S rRNA genes. Microbiome 6, 190.
Egozcue, J.J., Pawlowsky-Glahn, V., 2005. Groups of Parts and Their Balances in
Compositional Data Analysis. Mathematical Geology 37, 795–828.
El Aidy, S., Van Baarlen, P., Derrien, M., Lindenbergh-Kortleve, D.J., Hooiveld, G., Levenez, F.,
Doré, J., Dekker, J., Samsom, J.N., Nieuwenhuis, E.E., 2012. Temporal and spatial inter-
play of microbiota and intestinal mucosa drive establishment of immune homeostasis in
conventionalized mice. Mucosal Immunology 5, 567.
38 The Gut Microbiome: Bench to Table
Ellis, C.L., Ma, Z.-M., Mann, S.K., Li, C.-S., Wu, J., Knight, T.H., Yotter, T., Hayes, T.L.,
Maniar, A.H., Troia-Cancio, P.V., 2011. Molecular characterization of stool microbiota
in HIV-infected subjects by panbacterial and order-level 16S ribosomal DNA (rDNA)
quantification and correlations with immune activation. Journal of Acquired Immune
Deficiency Syndromes (1999) 57, 363.
Escudié, F., Auer, L., Bernard, M., Mariadassou, M., Cauquil, L., Vidal, K., Maman, S.,
Hernandez-Raquet, G., Combes, S., Pascal, G., 2017. FROGS: find, rapidly, OTUs with
galaxy solution. Bioinformatics 34, 1287–1294.
Fang, H., Huang, C., Zhao, H., Deng, M., 2015. CCLasso: correlation inference for composi-
tional data through Lasso. Bioinformatics 31, 3172–3180.
Faust, K., Sathirapongsasuti, J.F., Izard, J., Segata, N., Gevers, D., Raes, J., Huttenhower, C., 2012.
Microbial co-occurrence relationships in the human microbiome. PLoS Computational
Biology 8, e1002606.
Fernandes, A.D., Reid, J.N.S., Macklaim, J.M., McMurrough, T.A., Edgell, D.R., Gloor, G.B.,
2014. Unifying the analysis of high-throughput sequencing datasets: characterizing
RNA-seq, 16S rRNA gene sequencing and selective growth experiments by composi-
tional data analysis. Microbiome 2, 15.
Finstad, K.M., Probst, A.J., Thomas, B.C., Andersen, G.L., Demergasso, C., Echeverría, A.,
Amundson, R.G., Banfield, J.F., 2017. Microbial community structure and the persis-
tence of cyanobacterial populations in salt crusts of the Hyperarid Atacama Desert
from genome-resolved metagenomics. Frontiers in Microbiology 8, 1435.
Fisher, C.K., Mehta, P., 2014. Identifying keystone species in the human gut microbiome from
metagenomic timeseries using sparse linear regression. PLoS One 9, e102451.
Foster, Z.S.L., Sharpton, T.J., Grünwald, N.J., 2017. Metacoder: an R package for visualiza-
tion and manipulation of community taxonomic diversity data. PLOS Computational
Biology 13, e1005404.
Fox, J.G., Feng, Y., Theve, E.J., Raczynski, A., Fiala, J.L., Doernte, A.L., Williams, M.,
McFaline, J., Essigmann, J., Schauer, D., 2010. Gut microbes define liver cancer risk
in mice exposed to chemical and viral transgenic hepatocarcinogens. Gut 59, 88–97.
Fredriksson, N.J., Hermansson, M., Wilén, B.-M., 2014. Tools for T-RFLP data analysis using
Excel. BMC Bioinformatics 15, 361.
Freilich, S., Kreimer, A., Meilijson, I., Gophna, U., Sharan, R., Ruppin, E., 2010. The large-
scale organization of the bacterial network of ecological co-occurrence interactions.
Nucleic Acids Research 38, 3857–3868.
Friedman, J., Alm, E.J., 2012. Inferring correlation networks from genomic survey data.
PLoS Computational Biology 8, e1002687.
Gao, X., Huynh, B.-T., Guillemot, D., Glaser, P., Opatowski, L., 2018. Inference of significant
microbial interactions from longitudinal metagenomics data. Frontiers in Microbiology
9, 2319.
Ginige, M.P., Hugenholtz, P., Daims, H., Wagner, M., Keller, J., Blackall, L.L., 2004. Use of
stable-isotope probing, full-cycle rRNA analysis, and fluorescence in situ hybridiza-
tion-microautoradiography to study a methanol-fed denitrifying microbial community.
Applied and Environmental Microbiology 70, 588–596.
Gloor, G.B., Hummelen, R., Macklaim, J.M., Dickson, R.J., Fernandes, A.D., MacPhee, R.,
Reid, G., 2010. Microbiome profiling by illumina sequencing of combinatorial sequence-
tagged PCR products. PloS One 5, e15406.
Gloor, G.B., Macklaim, J.M., Pawlowsky-Glahn, V., Egozcue, J.J., 2017. Microbiome datasets
are compositional: and this is not optional. Frontiers in Microbiology 8, 2224.
Gonze, D., Coyte, K.Z., Lahti, L., Faust, K., 2018. Microbial communities as dynamical sys-
tems. Current Opinion in Microbiology 44, 41–49.
Green, M.R., Sambrook, J., 2012. Molecular cloning. A laboratory manual 4th. Cold Spring
Harbor Laboratory Press, New York.
Tools and Resources for Microbe Analysis 39
Guillamön, J.M., Sabaté, J., Barrio, E., Cano, J., Querol, A., 1998. Rapid identification of
wine yeast species based on RFLP analysis of the ribosomal internal transcribed spacer
(ITS) region. Archives of Microbiology 169, 387–392.
Guillou, L., Bachar, D., Audic, S., Bass, D., Berney, C., Bittner, L., Boutte, C., Burgaud, G.,
de Vargas, C., Decelle, J., Del Campo, J., Dolan, J.R., Dunthorn, M., Edvardsen, B.,
Holzmann, M., Kooistra, W.H., Lara, E., Le Bescot, N., Logares, R., Mahe, F.,
Massana, R., Montresor, M., Morard, R., Not, F., Pawlowski, J., Probert, I., Sauvadet, A.L.,
Siano, R., Stoeck, T., Vaulot, D., Zimmermann, P., Christen, R., 2013. The Protist
Ribosomal Reference database (PR2): a catalog of unicellular eukaryote small sub-
unit rRNA sequences with curated taxonomy. Nucleic Acids Research 41, D597–604.
Gužvić, M., 2013. The history of DNA sequencing/ISTORIJAT SEKVENCIRANJA DNK.
Journal of Medical Biochemistry 32, 301–312.
Hawinkel, S., Mattiello, F., Bijnens, L., Thas, O., 2017. A broken promise: microbiome
differential abundance methods do not control the false discovery rate. Briefings in
Bioinformatics 20, 210–221.
Heintz-Buschart, A., Wilmes, P., 2018. Human gut microbiome: function matters. Trends in
Microbiology 26, 563–574.
Hugenholtz, P., Tyson, G.W., Blackall, L.L., 2002. Design and evaluation of 16S rRNA-
targeted oligonucleotide probes for fluorescence in situ hybridization, Gene probes.
Humana Press, Totowa, NJ, pp. 29–42.
Iszatt, N., Janssen, S., Lenters, V., Dahl, C., Stigum, H., Knight, R., Mandal, S., Peddada, S.,
González, A., Midtvedt, T., Eggesbø, M., 2019. Environmental toxicants in breast milk
of Norwegian mothers and gut bacteria composition and metabolites in their infants at
1 month. Microbiome 7, 34.
Jackson, M.A., Bonder, M.J., Kuncheva, Z., Zierer, J., Fu, J., Kurilshikov, A., Wijmenga, C.,
Zhernakova, A., Bell, J.T., Spector, T.D., 2018. Detection of stable community struc-
tures within gut microbiota co-occurrence networks from different human populations.
PeerJournal 6, e4303.
Joossens, M., Huys, G., Cnockaert, M., De Preter, V., Verbeke, K., Rutgeerts, P., Vandamme, P.,
Vermeire, S., 2011. Dysbiosis of the faecal microbiota in patients with Crohn’s disease and
their unaffected relatives. Gut 60, 631–637.
Kageyama, S., Takeshita, T., Takeuchi, K., Asakawa, M., Matsumi, R., Furuta, M., Shibata, Y.,
Nagai, K., Ikebe, M., Morita, M., Masuda, M., Toh, Y., Kiyohara, Y., Ninomiya, T.,
Yamashita, Y., 2019. Characteristics of the salivary microbiota in patients with various
digestive tract cancers. Frontiers in Microbiology 10, 1780.
Kaul, A., Mandal, S., Davidov, O., Peddada, S.D., 2017. Analysis of microbiome data in the
presence of excess zeros. Frontiers in Microbiology 8, 2114–2114.
Klindworth, A., Pruesse, E., Schweer, T., Peplies, J., Quast, C., Horn, M., Glöckner, F.O.,
2012. Evaluation of general 16S ribosomal RNA gene PCR primers for classical and
next-generation sequencing-based diversity studies. Nucleic Acids Research 41, e1.
Koskinen, K., Pausan, M.R., Perras, A.K., Beck, M., Bang, C., Mora, M., Schilhabel,
A., Schmitz, R., Moissl-Eichinger, C., 2017. First insights into the diverse human
archaeome: specific detection of archaea in the gastrointestinal tract, lung, and nose
and on skin. MBio 8, e00824-17.
Kozich, J.J., Westcott, S.L., Baxter, N.T., Highlander, S.K., Schloss, P.D., 2013. Development
of a dual-index sequencing strategy and curation pipeline for analyzing amplicon
sequence data on the MiSeq Illumina sequencing platform. Applied and Environmental
Microbiology 79, 5112–5120.
Kuperman, A.A., Zimmerman, A., Hamadia, S., Ziv, O., Gurevich, V., Fichtman, B., Gavert, N.,
Straussman, R., Rechnitzer, H., Barzilay, M., 2019. Deep microbial analysis of mul-
tiple placentas shows no evidence for a placental microbiome. BJOG: An International
Journal of Obstetrics & Gynaecology 127, 159–169.
40 The Gut Microbiome: Bench to Table
Kurtz, Z.D., Müller, C.L., Miraldi, E.R., Littman, D.R., Blaser, M.J., Bonneau, R.A., 2015.
Sparse and Compositionally Robust Inference of Microbial Ecological Networks. PLoS
Computational Biology 11, e1004226.
Lam, H.Y., Clark, M.J., Chen, R., Chen, R., Natsoulis, G., O’huallachain, M., Dewey, F.E.,
Habegger, L., Ashley, E.A., Gerstein, M.B., 2012. Performance comparison of whole-
genome sequencing platforms. Nature Biotechnology 30, 78.
Langfelder, P., Horvath, S., 2008. WGCNA: an R package for weighted correlation network
analysis. BMC Bioinformatics 9, 559.
Langille, M.G.I., Zaneveld, J., Caporaso, J.G., McDonald, D., Knights, D., Reyes, J.A.,
Clemente, J.C., Burkepile, D.E., Vega Thurber, R.L., Knight, R., Beiko, R.G.,
Huttenhower, C., 2013. Predictive functional profiling of microbial communities using
16S rRNA marker gene sequences. Nature Biotechnology 31, 814–821.
Lê Cao, K.-A., Costello, M.-E., Lakis, V.A., Bartolo, F., Chua, X.-Y., Brazeilles, R., Rondeau, P.,
2016. MixMC: a multivariate statistical framework to gain insight into microbial com-
munities. PLoS One 11, e0160169.
Leaw, S.N., Chang, H.C., Sun, H.F., Barton, R., Bouchara, J.-P., Chang, T.C., 2006.
Identification of medically important yeast species by sequence analysis of the internal
transcribed spacer regions. Journal of Clinical Microbiology 44, 693–699.
LeBlanc, J.G., Milani, C., de Giori, G.S., Sesma, F., van Sinderen, D., Ventura, M., 2013.
Bacteria as vitamin suppliers to their host: a gut microbiota perspective. Current
Opinion in Biotechnology 24, 160–168.
Lee, I., Ouk Kim, Y., Park, S.C., Chun, J., 2016. OrthoANI: An improved algorithm and soft-
ware for calculating average nucleotide identity. International Journal of System and
Evolutionary Microbiology 66, 1100–1103.
Ley, R.E., 2010. Obesity and the human microbiome. Current Opinion in Gastroenterology
26, 5–11.
Li, R.W., Wu, S., Vi, R.L.B., Li, W., Li, C., 2012. Perturbation dynamics of the rumen micro-
biota in response to exogenous butyrate. PloS One 7, e29392.
Lim, K.M.K., Li, C., Chng, K.R., Nagarajan, N., 2016. @ MInter: automated text-mining of
microbial interactions. Bioinformatics 32, 2981–2987.
Liu, F., Wang, T.T.Y., Tang, Q., Xue, C., Li, R.W., Wu, V.C.H., 2019. Malvidin 3-glucoside
modulated gut microbial dysbiosis and global metabolome disrupted in a murine colitis
model induced by dextran sulfate sodium. Molecular Nutrition & Food Research 63,
e1900455.
Lo, C., Marculescu, R., 2017. MPLasso: inferring microbial association networks using prior
microbial knowledge. PLoS Computational Biology 13, e1005915.
Lo Presti, A., Zorzi, F., Del Chierico, F., Altomare, A., Cocca, S., Avola, A., De Biasio, F.,
Russo, A., Cella, E., Reddel, S., Calabrese, E., Biancone, L., Monteleone, G., Cicala, M.,
Angeletti, S., Ciccozzi, M., Putignani, L., Guarino, M.P.L., 2019. Fecal and mucosal
microbiota profiling in irritable bowel syndrome and inflammatory bowel disease.
Frontiers in Microbiology 10, 1655.
López-García, A., Pineda-Quiroga, C., Atxaerandio, R., Pérez, A., Hernández, I., García-
Rodríguez, A., González-Recio, O., 2018. Comparison of mothur and QIIME for the
analysis of rumen microbiota composition based on 16S rRNA amplicon sequences.
Frontiers in Microbiology 9, 3010.
Lozupone, C., Knight, R., 2005. UniFrac: a new phylogenetic method for comparing micro-
bial communities. Applied and Environmental Microbiology 71, 8228–8235.
Mandal, S., Van Treuren, W., White, R.A., Eggesbø, M., Knight, R., Peddada, S.D., 2015.
Analysis of composition of microbiomes: a novel method for studying microbial com-
position. Microbial Ecology in Health and Disease 26, 27663.
Marchesi, J.R., Ravel, J., 2015. The vocabulary of microbiome research: a proposal.
Microbiome 3, 1–3.
Tools and Resources for Microbe Analysis 41
Marsh, T.L., 1999. Terminal restriction fragment length polymorphism (T-RFLP): an emerg-
ing method for characterizing diversity among homologous populations of amplifica-
tion products. Current Opinion in Microbiology 2, 323–327.
McDonald, D., Price, M.N., Goodrich, J., Nawrocki, E.P., DeSantis, T.Z., Probst, A.,
Andersen, G.L., Knight, R., Hugenholtz, P., 2012. An improved Greengenes taxonomy
with explicit ranks for ecological and evolutionary analyses of bacteria and archaea.
ISME Journal 6, 610.
McHardy, I.H., Li, X., Tong, M., Ruegger, P., Jacobs, J., Borneman, J., Anton, P., Braun, J.,
2013. HIV Infection is associated with compositional and functional shifts in the rectal
mucosal microbiota. Microbiome 1, 26.
McMurdie, P.J., Holmes, S., 2014. Waste not, want not: why rarefying microbiome data is
inadmissible. PLOS Computational Biology 10, e1003531.
Meyer, F., Paarmann, D., D’Souza, M., Olson, R., Glass, E.M., Kubal, M., Paczian, T.,
Rodriguez, A., Stevens, R., Wilke, A., Wilkening, J., Edwards, R.A., 2008. The metage-
nomics RAST server – a public resource for the automatic phylogenetic and functional
analysis of metagenomes. BMC Bioinformatics 9, 386.
Mitra, S., Stärk, M., Huson, D.H., 2011. Analysis of 16S rRNA environmental sequences
using MEGAN. BMC Genomics 12, S17.
Morton, J.T., Sanders, J., Quinn, R.A., McDonald, D., Gonzalez, A., Vázquez-Baeza, Y.,
Navas-Molina, J.A., Song, S.J., Metcalf, J.L., Hyde, E.R., Lladser, M., Dorrestein, P.C.,
Knight, R., 2017. Balance trees reveal microbial niche differentiation. MSystems 2,
e00162-16.
Muyzer, G., De Waal, E.C., Uitterlinden, A.G., 1993. Profiling of complex microbial populations
by denaturing gradient gel electrophoresis analysis of polymerase chain reaction-amplified
genes coding for 16S rRNA. Applied and Environmental Microbiology 59, 695–700.
Muyzer, G., Smalla, K., 1998. Application of denaturing gradient gel electrophoresis (DGGE)
and temperature gradient gel electrophoresis (TGGE) in microbial ecology. Antonie
van Leeuwenhoek 73, 127–141.
Nakano, Y., Takeshita, T., Yamashita, Y., 2006. TRFMA: a web-based tool for termi-
nal restriction fragment length polymorphism analysis based on molecular weight.
Bioinformatics 22, 1788–1789.
Nelder, J.A., Mead, R., 1965. A simplex method for function minimization. The Computer
Journal 7, 308–313.
Nilsson, R.H., Larsson, K.-H., Taylor, A.F S., Bengtsson-Palme, J., Jeppesen, T.S., Schigel, D.,
Kennedy, P., Picard, K., Glöckner, F.O., Tedersoo, L., Saar, I., Kõljalg, U., Abarenkov,
K., 2018. The UNITE database for molecular identification of fungi: handling dark taxa
and parallel taxonomic classifications. Nucleic Acids Research 47, D259–D264.
Olesen, J.M., Bascompte, J., Dupont, Y.L., Jordano, P., 2007. The modularity of pollination
networks. Proceedings of the National Academy of Sciences of the United States of
America 104, 19891–19896.
Oliver, J.D., 2005. The viable but nonculturable state in bacteria. The Journal of Microbiology
43, 93–100.
Osborn, A.M., Moore, E.R., Timmis, K.N., 2000. An evaluation of terminal‐restriction frag-
ment length polymorphism (T‐RFLP) analysis for the study of microbial community
structure and dynamics. Environmental Microbiology 2, 39–50.
Pace, N.R., Stahl, D.A., Lane, D.J., Olsen, G.J., 1986. The analysis of natural microbial popu-
lations by ribosomal RNA sequences. In: Marshall, K.C. (eds) Advances in microbial
ecology. Springer, Boston, MA, pp. 1–55.
Pallmann, P., Schaarschmidt, F., Hothorn, L.A., Fischer, C., Nacke, H., Priesnitz, K.U.,
Schork, N.J., 2012. Assessing group differences in biodiversity by simultaneously test-
ing a user‐defined selection of diversity indices. Molecular Ecology Resources 12,
1068–1078.
42 The Gut Microbiome: Bench to Table
Paulson, J.N., Stine, O.C., Bravo, H.C., Pop, M., 2013. Differential abundance analysis for
microbial marker-gene surveys. Nature Methods 10, 1200–1202.
Peterson, C.T., Sharma, V., Iablokov, S.N., Albayrak, L., Khanipov, K., Uchitel, S., Chopra,
D., Mills, P.J., Fofanov, Y., Rodionov, D.A., Peterson, S.N., 2019. 16S rRNA gene profil-
ing and genome reconstruction reveal community metabolic interactions and prebiotic
potential of medicinal herbs used in neurodegenerative disease and as nootropics. PLoS
One 14, e0213869.
Pruesse, E., Quast, C., Knittel, K., Fuchs, B.M., Ludwig, W., Peplies, J., Glöckner, F.O., 2007.
SILVA: a comprehensive online resource for quality checked and aligned ribosomal
RNA sequence data compatible with ARB. Nucleic Acids Research 35, 7188–7196.
Puri, P., Liangpunsakul, S., Christensen, J.E., Shah, V.H., Kamath, P.S., Gores, G.J.,
Walker, S., Comerford, M., Katz, B., Borst, A., Yu, Q., Kumar, D.P., Mirshahi, F.,
Radaeva, S., Chalasani, N.P., Crabb, D.W., Sanyal, A.J., 2018. The circulating micro-
biome signature and inferred functional metagenomics in alcoholic hepatitis.
Hepatology 67, 1284–1302.
Quast, C., Pruesse, E., Yilmaz, P., Gerken, J., Schweer, T., Yarza, P., Peplies, J., Glöckner, F.O.,
2012. The SILVA ribosomal RNA gene database project: improved data processing and
web-based tools. Nucleic Acids Research 41, D590–D596.
Quinn, T.P., Erb, I., Gloor, G., Notredame, C., Richardson, M.F., Crowley, T.M., 2019. A field
guide for the compositional analysis of any-omics data. GigaScience 8, giz107.
Rhoads, A., Au, K.F., 2015. PacBio sequencing and its applications. Genomics, Proteomics &
Bioinformatics 13, 278–289.
Rivera-Pinto, J., Egozcue, J.J., Pawlowsky-Glahn, V., Paredes, R., Noguera-Julian, M., Calle, M.L.,
2018. Balances: a new perspective for microbiome analysis. MSystems 3, e00053-00018.
Rooks, M.G., Garrett, W.S., 2016. Gut microbiota, metabolites and host immunity. Nature
Reviews Immunology 16, 341–352.
Ruan, Q., Dutta, D., Schwalbach, M.S., Steele, J.A., Fuhrman, J.A., Sun, F., 2006. Local simi-
larity analysis reveals unique associations among marine bacterioplankton species and
environmental factors. Bioinformatics 22, 2532–2538.
Salipante, S. J., Kawashima, T., Rosenthal, C., Hoogestraat, D. R., Cummings, L. A.,
Sengupta, D. J., Harkins, T. T., Cookson, B. T., & Hoffman, N. G., 2014. Performance
comparison of Illumina and ion torrent next-generation sequencing platforms for 16S
rRNA-based bacterial community profiling. Applied and environmental microbiology
80, 7583–7591.
Sansonetti, P.J., Di Santo, J.P., 2007. Debugging how bacteria manipulate the immune
response. Immunity 26, 149–161.
Schaechter, M., 2015. A brief history of bacterial growth physiology. Frontiers in Microbiology
6, 289–289.
Schloss, P.D., Westcott, S.L., Ryabin, T., Hall, J.R., Hartmann, M., Hollister, E.B.,
Lesniewski, R.A., Oakley, B.B., Parks, D.H., Robinson, C.J., 2009. Introducing mothur:
open-source, platform-independent, community-supported software for describing
and comparing microbial communities. Applied and Environmental Microbiology 75,
7537–7541.
Schoch, C.L., Seifert, K.A., Huhndorf, S., Robert, V., Spouge, J.L., Levesque, C.A., Chen, W.,
Fungal Barcoding, C., Fungal Barcoding Consortium Author List, 2012. Nuclear ribo-
somal internal transcribed spacer (ITS) region as a universal DNA barcode marker
for fungi. Proceedings of the National Academy of Sciences of the United States of
America 109, 6241–6246.
Schwiertz, A., Jacobi, M., Frick, J.-S., Richter, M., Rusch, K., Köhler, H., 2010. Microbiota in
pediatric inflammatory bowel disease. The Journal of Pediatrics 157, 240–244.
Segata, N., Izard, J., Waldron, L., Gevers, D., Miropolsky, L., Garrett, W.S., Huttenhower, C.,
2011. Metagenomic biomarker discovery and explanation. Genome Biology 12, R60.
Tools and Resources for Microbe Analysis 43
Segata, N., Waldron, L., Ballarini, A., Narasimhan, V., Jousson, O., Huttenhower, C., 2012.
Metagenomic microbial community profiling using unique clade-specific marker genes.
Nat Methods 9, 811–814.
Sender, R., Fuchs, S., Milo, R., 2016. Revised estimates for the number of human and bacteria
cells in the body. PLoS Biolgy 14, e1002533–e1002533.
Shin, J., Lee, S., Go, M.-J., Lee, S.Y., Kim, S.C., Lee, C.-H., Cho, B.-K., 2016. Analysis of
the mouse gut microbiome using full-length 16S rRNA amplicon sequencing. Scientific
Reports 6, 29681.
Sims, T.T., Colbert, L.E., Zheng, J., Delgado Medrano, A.Y., Hoffman, K.L., Ramondetta, L.,
Jazaeri, A., Jhingran, A., Schmeler, K.M., Daniel, C.R., Klopp, A., 2019. Gut micro-
bial diversity and genus-level differences identified in cervical cancer patients versus
healthy controls. Gynecol Oncology 155, 237–244.
Soergel, D.A., Dey, N., Knight, R., Brenner, S.E., 2012. Selection of primers for optimal
taxonomic classification of environmental 16S rRNA gene sequences. ISME Journal
6, 1440.
Sommer, F., Bäckhed, F., 2013. The gut microbiota—masters of host development and physi-
ology. Nature Reviews Microbiology 11, 227.
Stevens, J.R., Jones, T.R., Lefevre, M., Ganesan, B., Weimer, B.C., 2017. SigTree: a
microbial community analysis tool to identify and visualize significantly respon-
sive branches in a phylogenetic tree. Computational and Structural Biotechnology
Journal 15, 372–378.
Stinson, L.F., Boyce, M.C., Payne, M.S., Keelan, J.A., 2019. The not-so-sterile womb: evidence
that the human fetus is exposed to bacteria prior to birth. Frontiers in Microbiology 10,
1124.
Theis, K.R., Romero, R., Winters, A.D., Greenberg, J.M., Gomez-Lopez, N., Alhousseini, A.,
Bieda, J., Maymon, E., Pacora, P., Fettweis, J.M., 2019. Does the human placenta deliv-
ered at term have a microbiota? Results of cultivation, quantitative real-time PCR,
16S rRNA gene sequencing, and metagenomics. American Journal of Obstetrics and
Gynecology 220, e261–e267.
Thijs, S., Op De Beeck, M., Beckers, B., Truyens, S., Stevens, V., Van Hamme, J.D., Weyens, N.,
Vangronsveld, J., 2017. Comparative evaluation of four bacteria-specific primer pairs
for 16S rRNA gene surveys. Frontiers in Microbiology 8, 494.
Tringe, S.G., Hugenholtz, P., 2008. A renaissance for the pioneering 16S rRNA gene. Current
Opinion in Microbiology 11, 442–446.
Trussart, M., Yus, E., Martinez, S., Baù, D., Tahara, Y.O., Pengo, T., Widjaja, M., Kretschmer, S.,
Swoger, J., Djordjevic, S., Turnbull, L., Whitchurch, C., Miyata, M., Marti-Renom, M.A.,
Lluch-Senar, M., Serrano, L., 2017. Defined chromosome structure in the genome-reduced
bacterium mycoplasma pneumoniae. Nature Communications 8, 14665.
Voelkerding, K.V., Dames, S.A., Durtschi, J.D., 2009. Next-generation sequencing: from basic
research to diagnostics. Clinical Chemistry 55, 641–658.
v. Wintzingerode, F., Göbel, U.B., Stackebrandt, E., 1997. Determination of microbial diversity
in environmental samples: pitfalls of PCR-based rRNA analysis. FEMS Microbiology
Reviews 21, 213–229.
Wang, J., Zheng, J., Shi, W., Du, N., Xu, X., Zhang, Y., Ji, P., Zhang, F., Jia, Z., Wang, Y.,
2018. Dysbiosis of maternal and neonatal microbiota associated with gestational diabe-
tes mellitus. Gut 67, 1614–1625.
Watts, S.C., Ritchie, S.C., Inouye, M., Holt, K.E., 2018. FastSpar: rapid and scalable correla-
tion estimation for compositional data. Bioinformatics 35, 1064–1066.
Weiss, S., Van Treuren, W., Lozupone, C., Faust, K., Friedman, J., Deng, Y., Xia, L.C., Xu, Z.Z.,
Ursell, L., Alm, E.J., Birmingham, A., Cram, J.A., Fuhrman, J.A., Raes, J., Sun, F.,
Zhou, J., Knight, R., 2016. Correlation detection strategies in microbial data sets vary
widely in sensitivity and precision. ISME Journal 10, 1669–1681.
44 The Gut Microbiome: Bench to Table
Weiss, S., Xu, Z.Z., Peddada, S., Amir, A., Bittinger, K., Gonzalez, A., Lozupone, C.,
Zaneveld, J.R., Vázquez-Baeza, Y., Birmingham, A., Hyde, E.R., Knight, R., 2017.
Normalization and microbial differential abundance strategies depend upon data char-
acteristics. Microbiome 5, 27.
Whitman, W.B., Coleman, D.C., Wiebe, W.J., 1998. Prokaryotes: the unseen majority.
Proceedings of the National Academy of Sciences 95, 6578–6583.
Wilks, J., Golovkina, T., 2012. Influence of microbiota on viral infections. PLoS Pathogens
8, e1002681.
Woese, C.R., 1987. Bacterial evolution. Microbiological Reviews 51, 221.
Woese, C.R., Fox, G.E., 1977. Phylogenetic structure of the prokaryotic domain: the primary
kingdoms. Proceedings of the National Academy of Sciences 74, 5088–5090.
Woese, C.R., Kandler, O., Wheelis, M.L., 1990. Towards a natural system of organisms:
proposal for the domains archaea, bacteria, and eucarya. Proceedings of the National
Academy of Sciences 87, 4576–4579.
Wu, I., Kim, H.S., Ben-Yehezkel, T., 2019. A single-molecule long-read survey of human tran-
scriptomes using LoopSeq synthetic long read sequencing. bioRxiv, 532135.
Yang, P., Yu, S., Cheng, L., Ning, K., 2019. Meta-network: optimized species-species network
analysis for microbial communities. BMC Genomics 20, 187.
Yoon, S.-H., Ha, S.-M., Kwon, S., Lim, J., Kim, Y., Seo, H., Chun, J., 2017. Introducing
EzBioCloud: a taxonomically united database of 16S rRNA gene sequences and
whole-genome assemblies. International Journal of Systematic and Evolutionary
Microbiology 67, 1613.
Zakrzewski, M., Proietti, C., Ellis, J.J., Hasan, S., Brion, M.-J., Berger, B., Krause, L., 2017.
Calypso: a user-friendly web-server for mining and visualizing microbiome–environment
interactions. Bioinformatics 33, 782–783.
2 Phageome in
Gut Microbiome
Yujie Zhang, Yen-Te Liao, Logan Tom,
Somanshu Sharma, and Vivian C.H. Wu
United States Department of Agriculture
CONTENTS
The Origins and Roles of Phages, Phageome, and Its Composition in
Human Gut Microbiota ............................................................................................ 45
How Do Phages Shape the Human Gut Microbiota ................................................ 49
Phage Interaction with the Immune System ............................................................ 52
Phageome and Health and Disease .......................................................................... 53
Phage-Based Therapy for Human Gut-Related Disease Is a Promising
Approach to Control/Prevent Human Disease ......................................................... 56
The Challenges of Phageome in Human Gut Microbiome, Perspectives,
and Future Directions............................................................................................... 59
References ................................................................................................................ 62
DOI: 10.1201/b22970-3 45
46 The Gut Microbiome: Bench to Table
The diversity and abundance of the gut microbiota are not simply present at
birth, but in humans, it develops in a newborn’s first 2–3 years of life and stabi-
lizes over adult years. The microbiota colonizes within the first 1–4 days of life,
where the diversity of the phage community is initially high, but a further steady
decrease in overall viral diversity in the first 2.5 years of life leads to a rise in
microbial populations and diversity. This eventually revives the viral population
through prophage induction, and these induced prophages in the human microbiota
become a crucial source of phages in the gut (Manrique et al., 2017). Adult phage
communities are relatively stable, and most phages at this time contain dsDNA
or ssDNA. A larger Microviridae to Caudovirales ratio has been correlated with
healthier adults, and most are temperate phages (Townsend et al., 2021). At the
point of development, the phage virome is also highly varied and sensitive to the
individual host gut microbiota, and for each individual, multiple different phages
are intertwined and play vital roles in the gut, which cannot be replicated in other
individual adults because of diet, environment, and maternal lineages (Hallowell
et al., 2021). Although the gut phage composition is mostly unique to the indi-
vidual, there does exist a reservoir of common phages, notably one example being
crAssphage, which was isolated in 73% of 450 global fecal metagenomes. Out of
4,301 phages isolated from two study individuals, 23 “core phages” were found in
more than 50% of healthy individuals around the world, questioning the composi-
tion of these core phages in the HGP and the role they play in health; it even has
been proposed that some of the core phages might play more impactful roles in
human health (Norman et al., 2015). In addition, it is still debated whether common
phages occur among people in the same environment, such as household members,
or among unrelated individuals, and evidence exists to support each view. The
details of the HGP are still poorly understood, but partially through predator–prey
relationships and lytic–lysogenic balances, an active phageome is key to a healthy
and functional gut ecosystem (Manrique et al., 2016).
Ogilvie et al. studied a human gut-specific phage named φB124-14, and phage
φB124-14’s genome was compared with 611 other human gut phages and 48 chro-
mosomal sequences from its host Bacteroides fragilis, which is a normal gut
microbe that is essential to healthy GI function (Ogilvie et al., 2012). Although
its role in the gut was not precisely defined, researchers found both phages and
bacteria in the gut are closely related in their genomes, possibly due to horizon-
tal gene transfer. Therefore, phages and microbes in the gut likely share a very
similar ecological niche. In addition, phage φB124-14 still plays a specialized role
through its thymidylate synthase gene, which is rare in gut viral genomes and
possibly helps with the fitness and function of B. fragilis through lysogeny. Since
B. fragilis is important in human mucosal immunity and nutrition, the findings
indicated that phage φB124-1 facilitates this healthy role of gut microbes in the
body by transferring essential genes between host cells. The finding by Stern et al.
supported the concept that specific phages or taxa of phages in a common phage
reservoir in the human population are correlated with human health. Researchers
identified 991 phages from the gut microbiota of 124 individuals using sequences
found in bacterial CRISPR genes, and 78% of these phages were present in two or
48 The Gut Microbiome: Bench to Table
more individuals (Stern et al., 2012). In his study, Stern et al. also observed varying
ratios of phages and bacteria, some individuals showing dominance in phages or
host bacteria, and although temperate phages dominate the phage reservoirs, there
are even different ratios of lytic and lysogenic phages, suggesting that the HGP
changes over a lifetime and varies among individuals. These findings provide a
glimpse into the complex ecology of gut microbiota, and more research is required
to fully understand the effect of phages.
The GI tract is a heterogeneous ecosystem that not only contains diverse organ-
isms but also distinguished sub-environments. Different locations in the gut respond
differently to food macromolecules, microorganisms, and pathogens, and each
organ in the tract is structured differently to fit its role in the digestive system. As a
result, distinct microbial communities can grow in different locations in the GI tract
(Bushman and Liang, 2021). Because most of the phage genome is still unknown and
the gut microbiota is naturally individualized, it is difficult to sample and characterize
phages in detail across the GI tract. Therefore, not many studies on the locations of
phages in the human gut exist. However, through animal models, several studies have
provided an overall profile of phages in the gut to shed more light and provide trends
in mammalian gut microbiota. Looft et al. compared the intestinal metagenomes of
pigs and found that the ileum displays a significant abundance of phages and phage-
related genes, and these numbers remained stable over the month-long study, sug-
gesting that phages are concentrated and consistent in the ileum (Looft et al., 2014).
Ileal bacteria, mostly consisting of facultative anaerobes like Streptococcaceae, and
nutrients that are absorbed in the ileum, such as vitamin B12, can make the ileum a
more comfortable location for phages to settle, compared to the colon (Hsu et al.,
2021). Because of physical barriers in the GI tract, such as the ileocecal valve, the
phage profile among organs can be different; despite sharing common phages, the
virome of the small intestine and the virome of the large intestine and rectum are
ultimately unique, which lends caution to the bias of fecal samples to study the gut
virome (Guerin and Hill, 2020).
Phages are also present at high numbers in the mucosa, which lines the epi-
thelial layer of the GI tract, and this limits pathogen colonization in the body.
Bao et al. added 8 log PFU/mL of phage PA13076 (isolated from chicken feces)
or BP96115 (induced from Salmonella Pullorum Spu-109) into mouse models for
31 days to track phages throughout the GI tract; they observed that phages gradu-
ally increase toward the end of the GI tract in mice, with the highest phage titer
(close to 6.5 log PFU/g) present in feces (Bao et al., 2020). In their study, lytic
phages are dormant from the stomach to the jejunum and begin to increase in the
ileum and colon reaching their peak in feces, while lysogenic phages maintain
relatively higher titers from the stomach to the colon but are highest in feces and
the cecum. Generally, phages do not settle in the location of the GI tract that is the
active site of digestion, such as the stomach and duodenum, because of the hydro-
chloric acid and proteolytic enzymes present, respectively. Overall, the specific
profile of the phage gut community in various GI organs still requires research,
but studies already show that phages inhabit distinct areas in the gut based on their
structures and function in the body.
Phageome in Gut Microbiome 49
lysogenic phages allow for coexistence by integrating with their host (Townsend
et al., 2021). The piggyback-the-winner dynamic is predominant in the absence of
pathogens and plays a crucial role in the genetic exchange. Prophages have shown up
to 5% of the functions in the human gut microbiota by sharing metabolism genes that
contribute to nutrient cycling and community stability, thus regulating cell growth
and competition (Oh et al., 2019b). Shkoporov and Hill reported the large titer of
lysogenic phages in human guts compared with lytic phages. Even in a high con-
centration of hosts, phages will choose to replicate with bacterial hosts (Shkoporov
and Hill, 2019). The gut microbiota abundantly adheres to the top mucin layer in
the GI tract where more temperate phages are found and piggyback-the-winner is
favored. Despite the risk of lysis, the gut microbiota benefits from phages in the GI
tract through lysogeny and regulation of colonization, and phages have formed an
important ecological niche with GI microbes.
From the discovery of core phages, such as crAssphage, different phages have
been correlated with healthy and ill individuals, posing the existence of the HGP,
a gut virome significantly correlated to human health. The disruption of this core
virome from equilibrium is either the cause or result of disease, so more investiga-
tion is required. Studying phages in relation to human health and gut microbiota
requires an overarching view of the GI ecosystem, which is still not well understood
(Garmaeva et al., 2019). One angle to this problem is studying the immune system.
To the human body, phages are foreign nonself entities that cross the gut epithelium
via transcytosis or phagocytosis and come into contact with innate immune cells,
such as macrophages and dendritic cells, eventually inducing an antiphage antibody
response in lymph nodes (Carroll-Portillo and Lin, 2019). The questions arise when
these interactions induce inflammation and immune signaling seen in responses
triggered by animal viruses; whether the interactions are beneficial or harmful
to the gut microbiota, it is yet clear that the ecological role of phages is tied to
human health. A phage-induced immune response can fight off a bacterial infection,
subsequently disrupting gut microbes. Phages in the mucosal layers modulate the
immune system likely through activating anti-inflammatory cytokines in the lam-
ina propria and suppressing inflammation in the gut, thus stabilizing the intestinal
environment and inhibiting nonspecific immune responses present in autoimmu-
nity, hypersensitivity, and infection in the GI tract (Carroll-Portillo and Lin, 2019;
Łusiak-Szelachowska et al., 2017).
Changes in the gut phageome have also been associated with diet, and it is well
established that diet has a large influence on gut microbiota. Food macromol-
ecules, such as proteins and digestible and nondigestible carbohydrates, induce
shifts in the gut microbiota, and subsequently affect overall diversity of gut micro-
biota (Singh et al., 2017). A study conducted by Townsend et al. had shown that
diet also impacts the structure and composition of the gut phageome (Townsend
et al., 2021). differences in diet linked with gut phageome differences have been
observed over the development of an individual human. Breast milk is high in fats,
and studies with mouse models with high-fat diets show increased Microviridae to
Siphoviridae ratios in the gut (Schulfer et al., 2020). As babies wean and eat solid
foods, phage composition continues to alter, and an increase in Pectobacterium
phages and a decrease in Lactobacillus phages and Streptococcus phages are seen
Phageome in Gut Microbiome 51
in adulthood. In addition, certain foods are richer sources of phages; fresh meats
and fermented foods, such as cheese, soybeans, and sauerkraut, are rich source
of phages, so diets concerning these foods may contribute to a wider and larger
gut phageome (Chukeatirote et al., 2018). Malnourishment, especially in infancy,
significantly disrupts a healthy phageome, which suggests that infancy is a crucial
period for phageome development. Malnourished youth contain a phageome that
is correlated with individuals with growth stunts and various gut illnesses (Reyes
et al., 2015).
Once the phageome stabilizes in adulthood, the individual diet has less of an effect
on gut phages; however, the diets from geographical communities and ethnic cultures
begin to play a larger role. Zuo et al. studied the gut phage DNA virome in 930
healthy individuals in Hong Kong and Yunnan, China, who spanned various ethnici-
ties and residencies, and geography played the most significant role in shaping both
individual’s gut virome and bacteriome (Zuo et al., 2020). It is not clear whether the
virome affects the bacteriome or vice versa, and geography is impacted by many fac-
tors, such as available food sources and region-specific food preparations. However,
certain diets in these regions that have been ethnically established, such as barley,
buttermilk tea, and Pu’er tea, contributed to virome differences. Urban and rural
residencies also affect gut virome; variations in personal hygiene and cleanliness in
the kitchen were associated with varying levels of viral diversity. Even though the
gut phage community and its regulation are still not fully understood, these studies
show the significant effect of environmental factors, diet in particular, on the human
gut phageome and bacteriome. Specifically, several foods have been shown to have a
direct effect on phages in the human gut, and by extension, gut bacteria as well. Oh
et al. reported that dietary fructose and short-chain fatty acids (SCFAs) promote the
induction of prophages in gut symbiont Lactobacillus reuteri in mouse models in a
RecA-dependent manner (Oh et al., 2019a). Researchers observed L. reuteri reducing
fructose from the diet to mannitol, and other gut microbes were able to convert man-
nitol to SCFA, which leads to prophage induction. This implies that diet affects gut
microbes, which alter the phageome; the phageome then alters gut bacteria by lysis
of lysogens. In the colon specifically, nutrient starvation was shown to drive phage
production. This likely has to be a consequence of changes in the gut microbiota
density and competition, possibly linked with the activation of the SOS response.
Garmaeva et al. studied phages in the guts of 11 healthy adults on a gluten-free diet
(GFD) and found a decrease of crAss-like phages, Microviridae, and Podoviridae
phages in GFD individuals (Garmaeva et al., 2021). Additionally, the comparison
of Bray–Curtis distances between the individuals with GFD and gluten-containing
diets revealed the change of the gut virome. Whether the addition of gluten can
reverse these changes was not studied, but researchers were able to see how changes
in the diet result in changes in the phageome. Cronin et al. researched the effects of
physical exercise and regular whey protein supplements on the human gut micro-
biota (Cronin et al., 2018). Out of 90 study individuals, most experiment participants
receiving whey protein experienced higher taxonomic richness and β-diversity in
their gut virome than control participants. Overall, diet significantly influences the
phages and bacteria in the human gut microbiota, although researchers are still try-
ing to understand mechanistic details.
52 The Gut Microbiome: Bench to Table
TABLE 2.1
Relationship between Gut Phageome and Human Diseases
Diseases Changes in the Gut Phageome Example References
Ulcerative colitis A decrease in diversity and richness of Caudovirales Zuo et al. (2019)
phages; an increase in abundance of Caudovirales
phages and Enterobacteria phages in particular
Each disease type and cohort have unique Norman et al. (2015)
bacteriophages; decreased bacterial diversity and
richness was inversely correlated to gut phageome
changes
Crohn’s disease A significant expansion of Caudovirales Norman et al. (2015)
bacteriophages
Temperate bacteriophages dominate the gut virome Clooney et al. (2019)
Decreased prevalence of core bacteriophages (phages Manrique et al. (2016)
preset in more than 20% of healthy individuals)
Largest abundance of phages in CD ileum tissue and Wagner et al. (2013)
CD gut wash sample
Type 1 diabetes Increase in the E. coli phage/E. coli ratio due to Tetz et al. 2019
prophage induction
Evenness of two major phage groups, Myoviridae and Townsend et al. (2021)
Podoviridae, was lower; diversity of intestinal E. coli
phages is significantly higher
Type 2 diabetes The relative numbers of the Myoviridae, Podoviridae, Ma et al. (2018)
Siphoviridae, and unclassified Caudovirales families
increased significantly
Abundance of phages specific to Enterobacteriaceae Chen et al. (2021)
hosts increased significantly
Acquired Increase in adenoviruses and viruses from Monaco et al. (2016)
immunodeficiency the Anelloviridae
syndrome (AIDS)
Stunting Lower phage diversity; a decrease in temperate phages Mirzaei et al. (2020)
was an increase in adenoviruses and viruses from the Anelloviridae that were associ-
ated with the HIV infection (Monaco et al., 2016). These studies thus solidify the link
between the gut phageome and the emergence of a variety of diseases.
Importantly, while there is limited information on the gut phageome, several stud-
ies indicated that most viral populations could not be assigned taxonomy and were
kept unknown as the viral “dark matter”. Finkbeiner et al. studied diarrhea samples
from 12 children and employed a novel “micro-mass sequencing” technique to detect
the presence of both known and unknown viruses in the samples (Finkbeiner et al.,
2008). The results showed that there were several known viruses in these samples,
such as rotaviruses, caliciviruses, astroviruses, and adenoviruses. Additionally, the
samples revealed the presence of several unknown viruses, which held very little
sequence similarity to viromes in the GenBank database. What this study shows
is that viruses are highly interconnected to gastrointestinal health, and understand-
ing and characterizing viral genomes can help lead to a broader understanding of
56 The Gut Microbiome: Bench to Table
both causes and treatments of prevalent diseases. Recently, several studies investi-
gated different techniques to explore this viral dark matter (Fitzgerald et al., 2021).
For instance, Benler et al. discussed their search for unknown human gut phages
using phage hallmark genes. Their studies led them to discover 3,738 complete phage
genomes from 451 different genera, thus illustrating how vast and unknown the
human gut phageome truly is (Benler et al., 2021). In the future, further studies and
techniques are needed to help explore this unknown gut phageome.
Contemporary phage therapy has relied on lytic phages and usually involves mul-
tiple types of phages to create “phage cocktails” for greater efficacy. As of today,
according to a review by Lin et al., phage therapy products have not been approved
for human use in the EU or the USA (Lin et al., 2017). However, phage therapy has
been approved and widely used in the biocontrol of foodborne pathogens by the food
industry (Kazi and Annapure, 2016; Niu et al., 2009). Research in the field of phage
therapy is still persistent, and the medical potential of phage therapy as an antibacte-
rial treatment is being studied globally. Work by Maura et al. tested the efficacy of
a three-phage cocktail (CLB_P1, CLB_P2, and CLB_P3) in eliminating enteroag-
gregative Escherichia coli (EAEC) O104:H4 55989Str strain using mouse models
(Maura et al., 2012). The results showcased that after 24 hours, the bacteriophage
treatment led to significantly lower concentrations of 55989Str in the ileum of the
mice and slightly lower concentrations in the fecal matter. Thus, the efficacy and
potential application of phages in eliminating pathogenic bacteria in the gut are evi-
dent. These decreases were only transient but with a return to baseline concentrations
of 55989Str 3 days after treatment. The results of bacterial regrowth after 3 days high-
light the limitations of contemporary phage therapy, indicating that more research is
needed to design long-lasting treatments against bacterial pathogens. On the other
hand, phage therapy has the potential as a supplemental treatment to aid other thera-
peutic procedures. One of the primary medical areas that phage-based therapy has
been tested on is cancer. Particularly, phage therapy has been explored to use in
combination with other conventional cancer therapies, like chemotherapy. Research
conducted by Zheng et al. studied mouse models with colorectal cancer to test the
ability of phages to aid chemotherapy in treating cancer (Zheng et al., 2019). The
bacteria Fusobacterium nucleatum tends to promote tumors along the gastrointesti-
nal tract, leading to colorectal cancer. Thus, the researchers used irinotecan-loaded
dextran nanoparticles covalently linked to azide-modified phages to selectively elim-
inate harmful Fusobacterium nucleatum in the gut. Their results revealed that this
phage-based treatment led to significantly more successful first-line chemotherapy
treatments in mice compared to controls. Additionally, the authors repeated the treat-
ment in piglets and found that there were no significant changes in hemocyte counts,
immunoglobulin and histamine levels, and liver and renal functions. The finding
suggests that phage-based therapy can potentially be used as a supplemental treat-
ment for colorectal cancer with little to no side effects.
In addition, there has been an interest in using phage therapy to deliver CRISPR-
Cas9 systems to the gut in order to make genomic edits for the microbiome. Lam
et al. used mouse models to study the effects of using engineered bacteriophages
as vehicles for CRISPR delivery into the gut (Lam et al., 2021). The researchers
engineered the filamentous bacteriophage M13 to deliver exogenous CRISPR-Cas9
DNA to Escherichia coli populations of the mouse GI tract. The results reported that
using phages as a delivery mechanism for CRISPR systems induced chromosomal
deletions in the selected bacteria in both in vitro and in vivo settings. However, some
conclusions showcased potential drawbacks of this technique, including bacterial
deletion of the CRISPR DNA to escape targeting. This study provides a proof of con-
cept for future studies and suggests that manipulating the gut microbiome is possible
by using a phage delivery system.
Phageome in Gut Microbiome 59
Yet another important benchwork tool for phage therapy applications to the human
gut is fecal viral transplants. Fecal viral transplants involve screening and obtaining
viral populations of stool samples from one healthy individual and transplanting the
viruses into the colon of another recipient individual (Bojanova and Bordenstein,
2016). Essentially, it allows the microbiota of the healthy individual to repopulate
the gut of the sick individual. This technique has been proposed as a therapeutic
treatment for gastrointestinal diseases, given the connection between gut microbiota
and human health and the role that phages play in this connection. One of the pri-
mary advantages of using fecal viral transplants is preventing and treating bacterial
infections of the gut. As described earlier, numerous pathogenic bacterial species
can disrupt the healthy gut microbiome and cause dysbiosis. Thus, it is important to
eliminate these pathogenic agents to restore a healthy gut. The use of bacteriophages
emerges as a possible solution. Zuo et al. compared the virome and bacterial microbi-
ome changes in Clostridium difficile infection (CDI) of the gut subjects treated with
fecal microbiota transplantation (FMT) or with vancomycin (Zuo et al., 2018). The
results showed that the FMT treatment significantly decreased Caudovirales DNA
in subjects with CDI. Additionally, FMT resulted in changes in both bacterial micro-
biota and virome in the gut, while the vancomycin treatment only changed the bacte-
rial composition of the gut. These findings suggest that FMT may be a beneficial way
to treat bacterial infection of the gut. Some applications of fecal viral transplantation
are still being tested in animal models. One such application is the use of fecal viral
transplantation in improving the restoration of the normal bacterial gut microbiota
after antibiotic treatment. Draper et al. disrupted the mice’s gut microbiome using
a combination of penicillin and streptomycin (Draper et al., 2020). After that, the
bacteriome of mice from either fecal viral transplants or fecal viral transplants with
heat and nuclease treatment (both of which killed the bacteriophages as controls)
was observed. The results indicated that the fecal viral transplanted mice showcased
a higher degree of resemblance to the pre-antibiotic treatment bacteriome. Moreover,
analysis of the gut viromes of both groups of mice demonstrated that the fecal viral
transplanted mice maintained the phages used in the transplantation over time, sug-
gesting long-term benefits. Thus, the results of this study suggest the role of fecal
viral transplantation is not only a primary treatment but also a way to mitigate side
effects and improve recovery after other medical treatments, such as antibiotics.
patients received significant clinical improvement, even total resolution of the infec-
tion (Clarke et al., 2020). Although localized pain and swelling were found in some
patients, researchers concluded that the phages were contaminated with endotox-
ins from raw phage lysates. Beatrix and Domingo-Calap reviewed similar results
concerning phage therapy clinical trials in GI diseases (Gutiérrez and Domingo-
Calap, 2020). Based on a randomized, double-blind, placebo-controlled trial treating
chronic diarrhea patients with a four-phage cocktail, patients experienced alleviated
symptoms after 28 days of oral treatment. This was correlated with reduced proin-
flammatory E. coli and reductions in inflammatory cytokines and enzymes, such
as aminotransferases. Alpha and beta diversity parameters were maintained, also
meaning the phages did not disrupt the gut microbiota. Phage therapy has been suc-
cessfully administered throughout clinical experiences, likely indicating the safety
and efficacy of phage therapy against GI diseases caused by pathogenic bacteria.
When studying phages in the human gut microbiota, there are still research gaps
and challenges. In general, some phage genomes are unknown and share little homol-
ogy with known phage sequences. Unknown and hypothetical viral sequences are
continually prevalent in phage research that often force current genomic and taxo-
nomic systems to think instead of seeing current data in a new light (Shkoporov
and Hill, 2019). Phages are constantly evolving, and the diversity of viral genomes
also hinders how well researchers can align phages with references. Metagenomic
approaches have successfully facilitated the characterization of different phages in
the environment and closely linked the gut virome and bacteriome; however, at this
time, pure genomic studies are not able to detail specific phage–host interactions.
As genomic and sequence-based tools become more popular, researchers will be
losing whole picture if the information from in vitro and in vivo studies is missing.
This would create a methodological bias that does not account for phage biology
and evolutionary history and low sensitivity toward small DNA yields from phages.
Moreover, unlike bacteria, phages lack universal phylogenetic markers, and thus, it is
very complicated to gather large quantities of viral genetic material within metage-
nomic data without contamination of bacterial sequences based on current protocols
(Garmaeva et al., 2019).
When utilizing phages in therapy or application, the stability of phages is also a
challenge because it is frequently variable across phage species and formulations,
which makes it difficult for researchers to maintain phage titers through experiments
and clinical trials (Garmaeva et al., 2019). If not frozen or cooled down, phages will
spontaneously mutate over long periods of storage, which can impair the fitness of
phages and research data (Garmaeva et al., 2019). In addition, there is generally a
lack of quality and safety guidelines for preparing phages, especially for therapy.
Although there are strict regulations for pharmaceutical products, few standards have
been addressed specifically for phage research and application. Also, phage research
currently lacks a simple, fast, and high-throughput method to screen phages. The
techniques, including double-layer agar plates, real-time PCR, and flow cytometry,
commonly used for phage research are not easily compatible with all phages while
producing quick results (Pires et al., 2020). To efficiently continue phage therapy, it is
required to improve experimental methods, including methods in genomics, molecular
biology, and microbiology; standards of pharmacokinetics and pharmacodynamics of
Phageome in Gut Microbiome 61
phages also need to be investigated (Luong et al., 2020). The optimal routes of admin-
istration, dosage, and the appropriate diseases for phage therapy are also research
gaps. Moreover, the phage resistance has not been currently addressed and remains
a barrier to more effective research and phage therapy (Yang et al., 2020). Therefore,
there can be uncertainty among researchers, patients, and consumers, regarding the
effectiveness of phages and the arms race between phages and their bacterial hosts.
As phages in the gut microbiota continue to be studied, there will still be gaps to be
investigated in the future, such as the exploration of the viral dark matter.
While understanding the multitude of gastrointestinal diseases and other health
disorders that arise from the dysbiosis of the gut microbiota remains a goal for
microbiology research, there are still many current and future studies focusing on
manipulating the gut microbiome for therapeutic benefit, despite the challenges and
research gaps. Since the microbiome is inherently linked to a variety of immune and
metabolic responses, ways to manipulate the microbiome by enriching or changing
its composition and diversity can hold the key to therapeutic treatments for a variety
of health conditions. One of the emerging techniques to use the gut microbiome for
therapeutic benefit is fecal microbiota transplantation which has shown successful
therapy in IBD clinical trials (Oka and Sartor, 2020). Still, it is not always effec-
tive. There have been many documented cases of fecal microbiota transfer failures,
where the technique failed to eliminate pathogenic bacterial species or resulted in the
recurrence of gastrointestinal problems, like diarrhea (Allegretti et al., 2018). Recent
research has been using fecal viral transplants instead of whole fecal microbiota
transplants. There have been a few studies recently that have shown proof of the
concept of this technique. Of note, Lin et al. researched the effectiveness of trans-
planting only VLPs, many of which are bacteriophages, in mice compared to per-
forming whole fecal microbiota transplants (Lin et al., 2019). The goal was to test the
efficacy of both techniques to treat small intestinal bacterial overgrowth (SIBO). The
results revealed that the transplantation of the VLPs was as effective as normal FMT.
Using only bacteriophages has several advantages over traditional FMT. First, fecal
microbiota transplants have the risk of transferring potentially dangerous pathogenic
bacterial species from the donor to the recipient. This risk is eliminated with fecal
viral transplants, as no bacteria are transferred during this procedure. Additionally,
fecal viral transplants could theoretically lead to higher efficacy than FMT since
researchers can carefully select and screen for specific phages or viruses from fecal
samples. While fecal viral transplants certainly have their advantages, there are still
numerous ways to improve the technique. Future research should focus on improving
the selection and isolation of phages from fecal samples to yield the most efficacious
results. Additionally, while this technique has been tested in animal models, future
studies will need to be conducted with human trials to see whether these results can
truly be extrapolated for human therapeutic benefit.
While fecal viral transplants can help add or eliminate bacterial species to the gut
microbiome, an emerging field of study focuses on the genetic manipulation of the
microbiome to combat diseases. One of the most interesting contemporary examples
of this has been using bacteriophages as vehicles to deliver CRISPR-Cas9 systems
to the gut, as previously mentioned. Additional research on CRISPR and phages
can potentially help revolutionize this field. There are several concerns regarding
62 The Gut Microbiome: Bench to Table
CRISPR and gene editing that must be taken into consideration and investigated by
future research. For example, one major topic of focus regarding current and future
studies is to reduce the off-target effects of the CRISPR-Cas9 system. Improving
specificity and screening of bacteriophages to be used as vehicles for the CRISPR
systems is also another potential topic for future research.
Overall, while several recent studies have already shown how effective under-
standing and manipulating the phageome of gut microbiota can be, there are many
mysteries in the field that remain to be discovered, as described above. Additionally,
there are many potential directions for the future of this expanding field, including
establishing core gut viral and genetic databases, studying the evolutionary adapta-
tions of the gut microbiota to changing viral environments, and observing specific
changes in host phenotypes in response to alterations in the gut phageome. Future
research will hopefully discover the underlying mechanism and interactions of gut
microbiota and find more efficient alternatives to these challenges related to human
health and diseases.
REFERENCES
Allegretti, J.R., Allegretti, A.S., Phelps, E., Xu, H., Fischer, M., Kassam, Z., 2018. Classifying
fecal microbiota transplantation failure: an observational study examining timing
and characteristics of fecal microbiota transplantation failures. Clin. Gastroenterol.
Hepatol. 16, 1832–1833. https://doi.org/10.1016/J.CGH.2017.10.031
Allen, H.K., Looft, T., Bayles, D.O., Humphrey, S., Levine, U.Y., Alt, D., Stanton, T.B., 2011.
Antibiotics in feed induce prophages in swine fecal microbiomes. MBio 2(6), e00260-
11. https://doi.org/10.1128/MBIO.00260-11/SUPPL_FILE/MBO006111199ST4.DOC
Altay, A., Yahiro, T., Bozdayi, G., Matsumoto, T., Sahin, F., Ozkan, S., Nishizono, A., Söderlund-
Venermo, M., Ahmed, K., 2015. Bufavirus Genotype 3 in Turkish children with
severe diarrhoea. Clin. Microbiol. Infect. 21, 965.e1–965.e4. https://doi.org/10.1016/j.
cmi.2015.06.006
Bäckhed, F., Ding, H., Wang, T., Hooper, L. V., Gou, Y.K., Nagy, A., Semenkovich, C.F.,
Gordon, J.I., 2004. The gut microbiota as an environmental factor that regulates fat stor-
age. Proc. Natl. Acad. Sci. U. S. A. 101, 15718. https://doi.org/10.1073/PNAS.0407076101
Bao, H., Zhang, H., Zhou, Y., Zhu, S., Pang, M., Shahin, K., Olaniran, A., Schmidt, S., Wang,
R., 2020. Transient carriage and low-level colonization of orally administrated lytic and
temperate phages in the gut of mice. Food Prod. Process. Nutr. 21(2), 1–8. https://doi.
org/10.1186/S43014-020-00029-7
Benler, S., Yutin, N., Antipov, D., Rayko, M., Shmakov, S., Gussow, A.B., Pevzner, P.,
Koonin, E. V., 2021. Thousands of previously unknown phages discovered in whole-
community human gut metagenomes. Microbiome 9(1), 1–17. https://doi.org/10.1186/
S40168-021-01017-W
Bojanova, D.P., Bordenstein, S.R., 2016. Fecal transplants: what is being transferred? PLoS
Biol. 14(7). https://doi.org/10.1371/JOURNAL.PBIO.1002503
Bushman, F., Liang, G., 2021. Assembly of the virome in newborn human infants. Curr. Opin.
Virol. 48, 17–22. https://doi.org/10.1016/J.COVIRO.2021.03.004
Camarillo-Guerrero, L.F., Almeida, A., Rangel-Pineros, G., Finn, R.D., Lawley, T.D., 2021.
Massive expansion of human gut bacteriophage diversity. Cell 184, 1098–1109.e9.
https://doi.org/10.1016/J.CELL.2021.01.029
Carroll-Portillo, A., Lin, H.C., 2019. Bacteriophage and the innate immune system: access and sig-
naling. Microorganisms 7(12), 625. https://doi.org/10.3390/MICROORGANISMS7120625
Phageome in Gut Microbiome 63
Chen, Q., Ma, X., Li, C., Shen, Y., Zhu, W., Zhang, Y., Guo, X., Zhou, J., Liu, C., 2021. Enteric
phageome alterations in patients with type 2 diabetes. Front. Cell. Infect. Microbiol.
856. https://doi.org/10.3389/fcimb.2020.575084
Cheng, L., Qi, C., Zhuang, H., Fu, T., Zhang, X., 2020. gutMDisorder: a comprehensive data-
base for dysbiosis of the gut microbiota in disorders and interventions. Nucleic Acids
Res. 48, D554–D560. https://doi.org/10.1093/NAR/GKZ843
Chukeatirote, E., Phongtang, W., Kim, J., Jo, A., Jung, L.S., Ahn, J., 2018. Significance of
bacteriophages in fermented soybeans: a review. Biomol. Concepts 9, 131–142. https://
doi.org/10.1515/BMC-2018-0012
Clarke, A.L., Soir, S. De, Jones, J.D., 2020. The safety and efficacy of phage therapy for bone
and joint infections: a systematic review. Antibiotics 9, 795. https://doi.org/10.3390/
ANTIBIOTICS9110795
Clooney, A.G., Sutton, T.D., Shkoporov, A.N., Holohan, R.K., Daly, K.M., O‘Regan, O., Ryan,
F.J., Draper, L.A., Plevy, S.E., Ross, R.P., Hill, C., 2019. Whole-virome analysis sheds
light on viral dark matter in inflammatory bowel disease. Cell Host Microbe 26(6),
764–778. https://doi.org/10.1016/j.chom.2019.10.009
Cronin, O., Barton, W., Skuse, P., Penney, N.C., Garcia-Perez, I., Murphy, E.F., Woods, T.,
Nugent, H., Fanning, A., Melgar, S., Falvey, E.C., Holmes, E., Cotter, P.D., O‘Sullivan, O.,
Molloy, M.G., Shanahan, F., 2018. A prospective metagenomic and metabolomic
analysis of the impact of exercise and/or whey protein supplementation on the gut
microbiome of sedentary adults. MSystems 3(3), e00044-18. https://doi.org/10.1128/
MSYSTEMS.00044-18
Dalmasso, M., Hill, C., Ross, R.P., 2014. Exploiting gut bacteriophages for human health.
Trends Microbiol. 22, 399–405. https://doi.org/10.1016/J.TIM.2014.02.010
Draper, L.A., Ryan, F.J., Dalmasso, M., Casey, P.G., McCann, A., Velayudhan, V., Ross, R.P.,
Hill, C., 2020. Autochthonous faecal viral transfer (FVT) impacts the murine micro-
biome after antibiotic perturbation. BMC Biol. 18(1), 1–14. https://doi.org/10.1186/
S12915-020-00906-0
Duerkop, B.A., Clements, C.V., Rollins, D., Rodrigues, J.L.M., Hooper, L.V., 2012. A com-
posite bacteriophage alters colonization by an intestinal commensal bacterium. Proc.
Natl. Acad. Sci. U. S. A. 109, 17621–17626. https://doi.org/10.1073/PNAS.1206136109/-/
DCSUPPLEMENTAL
Febvre, H.P., Rao, S., Gindin, M., Goodwin, N.D.M., Finer, E., Vivanco, J.S., Lu, S., Manter,
D.K., Wallace, T.C., Weir, T.L., 2019. PHAGE study: effects of supplemental bacterio-
phage intake on inflammation and gut microbiota in healthy adults. Nutrients 11(3),
666. https://doi.org/10.3390/NU11030666
Fernandes, M.A., Verstraete, S.G., Phan, T., Deng, X., Stekol, E., Lamere, B., Lynch, S.V.,
Heyman, M.B., Delwart, E., 2019. Enteric virome and bacterial microbiota in children
with ulcerative colitis and Crohn disease. J. Pediatr. Gastroenterol. Nutr. 68, 30–36.
https://doi.org/10.1097/MPG.0000000000002140
Finkbeiner, S.R., Allred, A.F., Tarr, P.I., Klein, E.J., Kirkwood, C.D., Wang, D., 2008.
Metagenomic analysis of human diarrhea: viral detection and discovery. PLoS Pathog.
4(2), e1000011. https://doi.org/10.1371/JOURNAL.PPAT.1000011
Fitzgerald, C.B., Shkoporov, A.N., Upadrasta, A., Khokhlova, E. V., Ross, R.P., Hill, C., 2021.
Probing the “Dark Matter” of the human gut phageome: culture assisted metagenomics
enables rapid discovery and host-linking for novel bacteriophages. Front. Cell. Infect.
Microbiol. 100. https://doi.org/10.3389/FCIMB.2021.616918
Frank, D.N., St. Amand, A.L., Feldman, R.A., Boedeker, E.C., Harpaz, N., Pace, N.R., 2007.
Molecular-phylogenetic characterization of microbial community imbalances in human
inflammatory bowel diseases. Proc. Natl. Acad. Sci. U. S. A. 104, 13780–13785. https://
doi.org/10.1073/PNAS.0706625104
64 The Gut Microbiome: Bench to Table
Fröhlich, E.E., Farzi, A., Mayerhofer, R., Reichmann, F., Jačan, A., Wagner, B., Zinser, E.,
Bordag, N., Magnes, C., Fröhlich, E., Kashofer, K., Gorkiewicz, G., Holzer, P., 2016.
Cognitive impairment by antibiotic-induced gut dysbiosis: analysis of gut microbiota-
brain communication. Brain. Behav. Immun. 56, 140–155. https://doi.org/10.1016/J.
BBI.2016.02.020
Gan, X.T., Ettinger, G., Huang, C.X., Burton, J.P., Haist, J. V., Rajapurohitam, V., Sidaway, J.E.,
Martin, G., Gloor, G.B., Swann, J.R., Reid, G., Karmazyn, M., 2014. Probiotic
administration attenuates myocardial hypertrophy and heart failure after myo-
cardial infarction in the rat. Circ. Heart Fail. 7, 491–499. https://doi.org/10.1161/
CIRCHEARTFAILURE.113.000978
Garmaeva, S., Gulyaeva, A., Sinha, T., Shkoporov, A.N., Clooney, A.G., Stockdale, S.R.,
Spreckels, J.E., Sutton, T.D.S., Draper, L.A., Dutilh, B.E., Wijmenga, C., Kurilshikov, A.,
Fu, J., Hill, C., Zhernakova, A., 2021. Stability of the human gut virome and effect of
gluten-free diet. Cell Rep. 35(7), 109132. https://doi.org/10.1016/J.CELREP.2021.109132
Garmaeva, S., Sinha, T., Kurilshikov, A., Fu, J., Wijmenga, C., Zhernakova, A., 2019. Studying
the gut virome in the metagenomic era: challenges and perspectives. BMC Biol. 17(1),
1–14. https://doi.org/10.1186/S12915-019-0704-Y
Gaykema, R.P.A., Goehler, L.E., Lyte, M., 2004. Brain response to cecal infection with
Campylobacter jejuni: analysis with Fos immunohistochemistry. Brain. Behav. Immun.
18, 238–245. https://doi.org/10.1016/J.BBI.2003.08.002
Grif, K., Dierich, M.P., Karch, H., Allerberger, F., 1998. Strain-specific differences in the
amount of Shiga toxin released from enterohemorrhagic Escherichia coli O157 fol-
lowing exposure to subinhibitory concentrations of antimicrobial agents. Eur. J. Clin.
Microbiol. Infect. Dis. 17, 761–766. https://doi.org/10.1007/S100960050181
Guerin, E., Hill, C., 2020. Shining light on human gut bacteriophages. Front. Cell. Infect.
Microbiol. 481. https://doi.org/10.3389/FCIMB.2020.00481
Gutiérrez, B., Domingo-Calap, P., 2020. Phage therapy in gastrointestinal diseases.
Microorganisms 8, 1–11. https://doi.org/10.3390/MICROORGANISMS8091420
Hallowell, H.A., Higgins, K. V., Roberts, M., Johnson, R.M., Bayne, J., Maxwell, H.S.,
Brandebourg, T., Hiltbold Schwartz, E., 2021. Longitudinal analysis of the intestinal
microbiota in the obese mangalica pig reveals alterations in bacteria and bacteriophage
populations associated with changes in body composition and diet. Front. Cell. Infect.
Microbiol. 934. https://doi.org/10.3389/FCIMB.2021.698657
Howard-Varona, C., Hargreaves, K.R., Abedon, S.T., Sullivan, M.B., 2017. Lysogeny in
nature: Mechanisms, impact and ecology of temperate phages. ISME Journal. 11(7),
1511–1520. https://doi.org/10.1038/ismej.2017.16
Hsu, B.B., Gibson, T.E., Yeliseyev, V., Liu, Q., Lyon, L., Bry, L., Silver, P.A., Gerber, G.K., 2019.
Dynamic modulation of the gut microbiota and metabolome by bacteriophages in a mouse
model. Cell Host Microbe 25, 803–814.e5. https://doi.org/10.1016/J.CHOM.2019.05.001
Hsu, C.L., Duan, Y., Fouts, D.E., Schnabl, B., 2021. Intestinal virome and therapeutic potential
of bacteriophages in liver disease. J. Hepatol. 75, 1465–1475. https://doi.org/10.1016/j.
jhep.2021.08.003
Kazi, M., Annapure, U.S., 2016. Bacteriophage biocontrol of foodborne pathogens. J. Food
Sci. Technol. 53(3), 1355–1362. https://doi.org/10.1007/s13197-015-1996-8
Keen, E.C., 2015. A century of phage research: bacteriophages and the shaping of modern
biology. Bioessays 37, 6. https://doi.org/10.1002/BIES.201400152
Kelly, J.R., Borre, Y., O’ Brien, C., Patterson, E., El Aidy, S., Deane, J., Kennedy, P.J.,
Beers, S., Scott, K., Moloney, G., Hoban, A.E., Scott, L., Fitzgerald, P., Ross, P.,
Stanton, C., Clarke, G., Cryan, J.F., Dinan, T.G., 2016. Transferring the blues:
depression-associated gut microbiota induces neurobehavioural changes in the rat. J.
Psychiatr. Res. 82, 109–118. https://doi.org/10.1016/J.JPSYCHIRES.2016.07.019
Phageome in Gut Microbiome 65
Kieft, K., Zhou, Z., Anderson, R.E., Buchan, A., Campbell, B.J., Hallam, S.J., Hess, M.,
Sullivan, M.B., Walsh, D.A., Roux, S., Anantharaman, K., 2021. Ecology of inorganic
sulfur auxiliary metabolism in widespread bacteriophages. Nat. Commun. 121(12),
1–16. https://doi.org/10.1038/s41467-021-23698-5
Kim, K.W., Horton, J.L., Pang, C.N.I., Jain, K., Leung, P., Isaacs, S.R., Bull, R.A., Luciani, F.,
Wilkins, M.R., Catteau, J., Lipkin, W.I., Rawlinson, W.D., Briese, T., Craig, M.E., 2019.
Higher abundance of enterovirus A species in the gut of children with islet autoimmu-
nity. Sci. Rep. 9(1), 1–8. https://doi.org/10.1038/S41598-018-38368-8
Lam, K.N., Spanogiannopoulos, P., Soto-Perez, P., Alexander, M., Nalley, M.J., Bisanz, J.E.,
Nayak, R.R., Weakley, A.M., Yu, F.B., Turnbaugh, P.J., 2021. Phage-delivered CRISPR-
Cas9 for strain-specific depletion and genomic deletions in the gut microbiome. Cell
Rep. 37(5), 109930. https://doi.org/10.1016/J.CELREP.2021.109930
Lam, V., Su, J., Hsu, A., Gross, G.J., Salzman, N.H., Baker, J.E., 2016. Intestinal microbial
metabolites are linked to severity of myocardial infarction in rats. PLoS One 11(8),
e0160840. https://doi.org/10.1371/JOURNAL.PONE.0160840
Letarov, A.V., 2020. History of early bacteriophage research and emergence of key con-
cepts in virology. Biochemistry (Mosc.) 85, 1093–1112. https://doi.org/10.1134/
S0006297920090096
Lewis, R., Hill, C., 2020. Overcoming barriers to phage application in food and feed. Curr.
Opin. Biotechnol. 61, 38–44. https://doi.org/10.1016/j.copbio.2019.09.018
Ley, R.E., Bäckhed, F., Turnbaugh, P., Lozupone, C.A., Knight, R.D., Gordon, J.I., 2005.
Obesity alters gut microbial ecology. Proc. Natl. Acad. Sci. U. S. A. 102(31), 11070–
11075. https://doi.org/10.1073/pnas.0504978102
Lin, D.M., Koskella, B., Lin, H.C., 2017. Phage therapy: an alternative to antibiotics in the age
of multi-drug resistance. World J. Gastrointest. Pharmacol. Ther. 8, 162. https://doi.
org/10.4292/WJGPT.V8.I3.162
Lin, D.M., Koskella, B., Ritz, N.L., Lin, D., Carroll-Portillo, A., Lin, H.C., 2019. Transplanting
fecal virus-like particles reduces high-fat diet-induced small intestinal bacterial over-
growth in mice. Front. Cell. Infect. Microbiol. 348. https://doi.org/10.3389/FCIMB.
2019.00348
Looft, T., Allen, H.K., Cantarel, B.L., Levine, U.Y., Bayles, D.O., Alt, D.P., Henrissat, B.,
Stanton, T.B., 2014. Bacteria, phages and pigs: the effects of in-feed antibiotics on the
microbiome at different gut locations. ISME J. 8, 1566–1576. https://doi.org/10.1038/
ISMEJ.2014.12
Luong, T., Salabarria, A.C., Roach, D.R., 2020. Phage therapy in the resistance era:
where do we stand and where are we going? Clin. Ther. 42, 1659–1680. https://doi.
org/10.1016/J.CLINTHERA.2020.07.014
Łusiak-Szelachowska, M., Weber-Dąbrowska, B., Jończyk-Matysiak, E., Wojciechowska, R.,
Górski, A., 2017. Bacteriophages in the gastrointestinal tract and their implications. Gut
Pathog. 9(1), 1–5. https://doi.org/10.1186/S13099-017-0196-7
Ma, Y., You, X., Mai, G., Tokuyasu, T. and Liu, C., 2018. A human gut phage catalog cor-
relates the gut phageome with type 2 diabetes. Microbiome 6(1), 1–12. https://doi.
org/10.1186/s40168-018-0410–y
Manrique, P., Bolduc, B., Walk, S.T., Der Van Oost, J., De Vos, W.M., Young, M.J., 2016. Healthy
human gut phageome. Proc. Natl. Acad. Sci. U. S. A. 113, 10400–10405. https://doi.
org/10.1073/PNAS.1601060113
Manrique, P., Dills, M., Young, M.J., 2017. The human gut phage community and its implica-
tions for health and disease. Viruses 9(6), 141. https://doi.org/10.3390/V9060141
Maura, D., Galtier, M., Le Bouguénec, C., Debarbieux, L., 2012. Virulent bacteriophages can
target O104:H4 enteroaggregative Escherichia coli in the mouse intestine. Antimicrob.
Agents Chemother. 56, 6235–6242. https://doi.org/10.1128/AAC.00602-12
66 The Gut Microbiome: Bench to Table
Mirzaei, M.K., Khan, M.A.A., Ghosh, P., Taranu, Z.E., Taguer, M., Ru, J., Chowdhury, R.,
Kabir, M.M., Deng, L., Mondal, D., Maurice, C.F., 2020. Bacteriophages isolated from
stunted children can regulate gut bacterial communities in an age-specific manner. Cell
Host Microbe 27(2), 199–212. https://doi.org/10.1016/j.chom.2020.01.004
Monaco, C.L., Gootenberg, D.B., Zhao, G., Handley, S.A., Ghebremichael, M.S., Lim, E.S.,
Lankowski, A., Baldridge, M.T., Wilen, C.B., Flagg, M., Norman, J.M., Keller, B.C.,
Luévano, J.M., Wang, D., Boum, Y., Martin, J.N., Hunt, P.W., Bangsberg, D.R.,
Siedner, M.J., Kwon, D.S., Virgin, H.W., 2016. Altered virome and bacterial micro-
biome in human immunodeficiency virus-associated acquired immunodeficiency syn-
drome. Cell Host Microbe 19, 311–322. https://doi.org/10.1016/J.CHOM.2016.02.011
Mushegian, A.R., 2020. Are there 1031 virus particles on earth, or more, or fewer? J. Bacteriol.
202. https://doi.org/10.1128/JB.00052-20
Niu, Y.D., McAllister, T.A., Xu, Y., Johnson, R.P., Stephens, T.P., Stanford, K., 2009.
Prevalence and impact of bacteriophages on the presence of Escherichia coli O157:H7
in feedlot cattle and their environment. Appl. Environ. Microbiol. 75, 1271–1278.
https://doi.org/10.1128/AEM.02100-08
Norman, J.M., Handley, S.A., Baldridge, M.T., Droit, L., Liu, C.Y., Keller, B.C., Kambal, A.,
Monaco, C.L., Zhao, G., Fleshner, P., Stappenbeck, T.S., McGovern, D.P.B.,
Keshavarzian, A., Mutlu, E.A., Sauk, J., Gevers, D., Xavier, R.J., Wang, D., Parkes, M.,
Virgin, H.W., 2015. Disease-specific alterations in the enteric virome in inflammatory
bowel disease. Cell 160, 447. https://doi.org/10.1016/J.CELL.2015.01.002
Ogilvie, L.A., Caplin, J., Dedi, C., Diston, D., Cheek, E., Bowler, L., Taylor, H., Ebdon, J.,
Jones, B.V., 2012. Comparative (meta)genomic analysis and ecological profiling of
human gut-specific bacteriophage φB124-14. PLoS One 7, 35053. https://doi.org/10.1371/
JOURNAL.PONE.0035053
Oh, J.H., Alexander, L.M., Pan, M., Schueler, K.L., Keller, M.P., Attie, A.D., Walter, J., van
Pijkeren, J.P., 2019a. Dietary fructose and microbiota-derived short-chain fatty acids
promote bacteriophage production in the gut symbiont Lactobacillus reuteri. Cell Host
Microbe 25, 273–284.e6. https://doi.org/10.1016/J.CHOM.2018.11.016
Oh, J.H., Lin, X.B., Zhang, S., Tollenaar, S.L., Özçam, M., Dunphy, C., Walter, J., van
Pijkeren, J.P., 2019b. Prophages in Lactobacillus reuteri are associated with fitness
trade-offs but can increase competitiveness in the gut ecosystem. Appl. Environ.
Microbiol. 86(1), e01922-19. https://doi.org/10.1128/AEM.01922-19
Oka, A., Sartor, R.B., 2020. Microbial-based and microbial-targeted therapies for inflammatory
bowel diseases. Dig. Dis. Sci. 65, 757–788. https://doi.org/10.1007/S10620-020-06090-Z
Pires, D.P., Costa, A.R., Pinto, G., Meneses, L., Azeredo, J., 2020. Current challenges and
future opportunities of phage therapy. FEMS Microbiol. Rev. 44, 684–700. https://doi.
org/10.1093/FEMSRE/FUAA017
Reyes, A., Blanton, L. V., Cao, S., Zhao, G., Manary, M., Trehan, I., Smith, M.I., Wang, D.,
Virgin, H.W., Rohwer, F., Gordon, J.I., 2015. Gut DNA viromes of Malawian twins dis-
cordant for severe acute malnutrition. Proc. Natl. Acad. Sci. U. S. A. 112, 11941–11946.
https://doi.org/10.1073/PNAS.1514285112
Reyes, A., Haynes, M., Hanson, N., Angly, F.E., Heath, A.C., Rohwer, F., Gordon, J.I., 2010.
Viruses in the faecal microbiota of monozygotic twins and their mothers. Nature
466(7304), 334–338. https://doi.org/10.1038/nature09199
Ridaura, V.K., Faith, J.J., Rey, F.E., Cheng, J., Duncan, A.E., Kau, A.L., Griffin, N.W.,
Lombard, V., Henrissat, B., Bain, J.R., Muehlbauer, M.J., Ilkayeva, O., Semenkovich, C.F.,
Funai, K., Hayashi, D.K., Lyle, B.J., Martini, M.C., Ursell, L.K., Clemente, J.C.,
Van Treuren, W., Walters, W.A., Knight, R., Newgard, C.B., Heath, A.C., Gordon, J.I.,
Phageome in Gut Microbiome 67
2013. Cultured gut microbiota from twins discordant for obesity modulate adiposity
and metabolic phenotypes in mice. Science 341(6150), 1241214. https://doi.org/10.1126/
SCIENCE.1241214
Roux, S., Brum, J.R., Dutilh, B.E., Sunagawa, S., Duhaime, M.B., Loy, A., Poulos, B.T.,
Solonenko, N., Lara, E., Poulain, J., Pesant, S., Kandels-Lewis, S., Dimier, C., Picheral, M.,
Searson, S., Cruaud, C., Alberti, A., Duarte, C.M., Gasol, J.M., Vaqué, D., Bork, P.,
Acinas, S.G., Wincker, P., Sullivan, M.B., 2016. Ecogenomics and potential biogeo-
chemical impacts of globally abundant ocean viruses. Nature 537(7622), 689–693.
https://doi.org/10.1038/nature19366
Scanlan, P.D., 2020. Resistance may be futile: gut spatial heterogeneity supports bacteria-
phage co-existence. Cell Host Microbe 28, 356–358. https://doi.org/10.1016/J.CHOM.
2020.08.008
Schulfer, A., Santiago-Rodriguez, T.M., Ly, M., Borin, J.M., Chopyk, J., Blaser, M.J.,
Pride, D.T., 2020. Fecal viral community responses to high-fat diet in mice. mSphere
5(1), e00833-19. https://doi.org/10.1128/MSPHERE.00833-19
Seth, R.K., Maqsood, R., Mondal, A., Bose, D., Kimono, D., Holland, L.A., Lloyd, P.J.,
Klimas, N., Horner, R.D., Sullivan, K., Lim, E.S., Chatterjee, S., 2019. Gut DNA virome
diversity and its association with host bacteria regulate inflammatory phenotype and
neuronal immunotoxicity in experimental Gulf War illness. Viruses 11(10), 968. https://
doi.org/10.3390/V11100968
Shkoporov, A.N., Hill, C., 2019. Bacteriophages of the human gut: the “known unknown”
of the microbiome. Cell Host Microbe 25, 195–209. https://doi.org/10.1016/J.CHOM.
2019.01.017
Singh, R.K., Chang, H.W., Yan, D., Lee, K.M., Ucmak, D., Wong, K., Abrouk, M.,
Farahnik, B., Nakamura, M., Zhu, T.H., Bhutani, T., Liao, W., 2017. Influence of diet
on the gut microbiome and implications for human health. J. Transl. Med. 151(1), 1–17.
https://doi.org/10.1186/S12967-017-1175-Y
Sinha, A., Maurice, C.F., 2019. Bacteriophages: uncharacterized and dynamic regulators of
the immune system. Mediators Inflamm. https://doi.org/10.1155/2019/3730519
Stern, A., Mick, E., Tirosh, I., Sagy, O., Sorek, R., 2012. CRISPR targeting reveals a reser-
voir of common phages associated with the human gut microbiome. Genome Res. 22,
1985–1994. https://doi.org/10.1101/GR.138297.112
Sudo, N., Chida, Y., Aiba, Y., Sonoda, J., Oyama, N., Yu, X.N., Kubo, C., Koga, Y., 2004. Postnatal
microbial colonization programs the hypothalamic-pituitary-adrenal system for stress
response in mice. J. Physiol. 558, 263–275. https://doi.org/10.1113/JPHYSIOL.2004.063388
Sutcliffe, S.G., Shamash, M., Hynes, A.P., Maurice, C.F., 2021. Common oral medications
lead to prophage induction in bacterial isolates from the human gut. Viruses 13, 455.
https://doi.org/10.3390/V13030455
Tetz, G., Brown, S.M., Hao, Y., Tetz, V., 2019. Type 1 diabetes: an association between auto-
immunity, the dynamics of gut amyloid-producing E. coli and their phages. Sci. Rep.
9(1), 1–11. https://doi.org/10.1038/S41598-019-46087-X
Townsend, E.M., Kelly, L., Muscatt, G., Box, J.D., Hargraves, N., Lilley, D., Jameson, E.,
2021. The human gut phageome: origins and roles in the human gut microbiome. Front.
Cell. Infect. Microbiol. 498. https://doi.org/10.3389/FCIMB.2021.643214
Trubl, G., Jang, H. Bin, Roux, S., Emerson, J.B., Solonenko, N., Vik, D.R., Solden, L.,
Ellenbogen, J., Runyon, A.T., Bolduc, B., Woodcroft, B.J., Saleska, S.R., Tyson, G.W.,
Wrighton, K.C., Sullivan, M.B., Rich, V.I., 2018. Soil viruses are underexplored play-
ers in ecosystem carbon processing. mSystems 3(5), e00076-18. https://doi.org/10.1128/
mSystems.00076-18
68 The Gut Microbiome: Bench to Table
Wagner, J., Maksimovic, J., Farries, G., Sim, W.H., Bishop, R.F., Cameron, D.J., Catto-Smith,
A.G., Kirkwood, C.D., 2013. Bacteriophages in gut samples from pediatric Crohn’s
disease patients: metagenomic analysis using 454 pyrosequencing. Inflamm. Bowel Dis.
19(8), 1598–1608. https://doi.org/10.1097/MIB.0b013e318292477c
Yang, Y., Shen, W., Zhong, Q., Chen, Q., He, X., Baker, J.L., Xiong, K., Jin, X., Wang, J.,
Hu, F., Le, S., 2020. Development of a bacteriophage cocktail to constrain the emer-
gence of phage-resistant Pseudomonas aeruginosa. Front. Microbiol. 11, 327. https://
doi.org/10.3389/FMICB.2020.00327
Yinda, C.K., Vanhulle, E., Conceição-Neto, N., Beller, L., Deboutte, W., Shi, C., Ghogomu, S.M.,
Maes, P., Van Ranst, M., Matthijnssens, J., 2019. Gut virome analysis of cameroonians
reveals high diversity of enteric viruses, including potential interspecies transmitted
viruses. mSphere. 4(1), e00585-18. https://doi.org/10.1128/MSPHERE.00585-18
Zhang, Y., Liao, Y.-T. Te Salvador, A., Sun, X., Wu, V.C.H., 2020. Prediction, diversity, and
genomic analysis of temperate phages induced from shiga toxin-producing Escherichia
coli strains. Front. Microbiol. 10, 3093. https://doi.org/10.3389/fmicb.2019.03093
Zheng, D.W., Dong, X., Pan, P., Chen, K.W., Fan, J.X., Cheng, S.X., Zhang, X.Z., 2019. Phage-
guided modulation of the gut microbiota of mouse models of colorectal cancer aug-
ments their responses to chemotherapy. Nat. Biomed. Eng. 39(3), 717–728. https://doi.
org/10.1038/s41551-019-0423-2
Zhu, Q., Gao, R., Zhang, Y., Pan, D., Zhu, Y., Zhang, X., Yang, R., Jiang, R., Xu, Y., Qin, H., 2018.
Dysbiosis signatures of gut microbiota in coronary artery disease. Physiol. Genomics
50, 893–903. https://doi.org/10.1152/physiolgenomics.00070.2018
Zuo, T., Liu, Q., Zhang, F., Yeoh, Y.K., Wan, Y., Zhan, H., Lui, G.C.Y., Chen, Z., Li, A.Y.L.,
Cheung, C.P., Chen, N., Lv, W., Ng, R.W.Y., Tso, E.Y.K., Fung, K.S.C., Chan, V.,
Ling, L., Joynt, G., Hui, D.S.C., Chan, F.K.L., Chan, P.K.S., Ng, S.C., 2021. Temporal
landscape of human gut RNA and DNA virome in SARS-CoV-2 infection and severity.
Microbiome 9(1), 1–16. https://doi.org/10.1186/S40168-021-01008-X
Zuo, T., Lu, X.J., Zhang, Y., Cheung, C.P., Lam, S., Zhang, F., Tang, W., Ching, J.Y.L., Zhao, R.,
Chan, P.K.S., Sung, J.J.Y., Yu, J., Chan, F.K.L., Cao, Q., Sheng, J.Q., Ng, S.C., 2019.
Gut mucosal virome alterations in ulcerative colitis. Gut 68, 1169–1179. https://doi.
org/10.1136/GUTJNL-2018-318131
Zuo, T., Sun, Y., Wan, Y., Yeoh, Y.K., Zhang, F., Cheung, C.P., Chen, N., Luo, J., Wang, W.,
Sung, J.J.Y., Chan, P.K.S., Wang, K., Chan, F.K.L., Miao, Y., Ng, S.C., 2020. Human-
gut-DNA virome variations across geography, ethnicity, and urbanization. Cell Host
Microbe 28, 741–751.e4. https://doi.org/10.1016/J.CHOM.2020.08.005
Zuo, T., Wong, S.H., Lam, K., Lui, R., Cheung, K., Tang, W., Ching, J.Y.L., Chan, P.K.S.,
Chan, M.C.W., Wu, J.C.Y., Chan, F.K.L., Yu, J., Sung, J.J.Y., Ng, S.C., 2018. Bacteriophage
transfer during faecal microbiota transplantation in Clostridium difficile infec-
tion is associated with treatment outcome. Gut 67, 634–643. https://doi.org/10.1136/
GUTJNL-2017-313952
Section II
T1 Preclinical Animal
Studies, and Safety and
Efficacy Trial in Humans
3 Microbiome and Nutrition –
Why Do We Care?
Discussing a Complex
Relationship
Angelos K. Sikalidis
California Polytechnic State University
CONTENTS
Introductory Remarks .............................................................................................. 71
The Human Gut Microbiome ................................................................................... 74
The Metabolome and Microbiome Connection ....................................................... 75
Host–Gut Microbiome Metabolic Interactions ................................................... 76
Microbiome – Diet Interactions ............................................................................... 78
The Gut Microbiome and Carbohydrate Metabolism .............................................. 79
General Considerations ....................................................................................... 79
Simple Carbohydrates .........................................................................................80
Complex Carbohydrates ...................................................................................... 82
Nondigestible Polysaccharides – Fiber ............................................................... 82
The Gut Microbiome and Fatty Acid Metabolism ...................................................84
General Considerations .......................................................................................84
Saturated Fatty Acids .......................................................................................... 85
Polyunsaturated Fatty Acids (PUFAs) – Omega-3 and -6 (ω-3 and ω-6)
Fatty Acids .......................................................................................................... 86
Short-Chain Fatty Acids ...................................................................................... 88
The Gut Microbiome and Amino Acid Metabolism ................................................ 89
The Microbiome and Weight Change ......................................................................90
Future Perspectives ..................................................................................................92
References ................................................................................................................92
INTRODUCTORY REMARKS
The term microbiome arises from the Greek words “μικρο” – micro meaning “small”
(in mathematics and sciences designated with the Greek letter μ (10 −6)) – “βίωμα” –
biome meaning “of life”, in Greek: “μικροβίωμα”. Prior to 2001, the term micro-
biome was in use, mostly to infer a rather small significantly complex ecological
niche incorporating plant and animal life. While the study of what is now known as
DOI: 10.1201/b22970-5 71
72 The Gut Microbiome: Bench to Table
the “human microbiome” can be traced as far back as Antonie van Leeuwenhoek
(1632–1723), “I then most always saw, with great wonder, that in the said matter there
were many very little living animalcules, very prettily a-moving”. Leeuwenhoek,
who is known to have made over 500 “microscopes”, of which fewer than ten have
survived to the present day, was a keen, laborious and systematic observer of the
microbial world.
In 2001, the term “microbiome” was re-discovered, re-surfaced and used by Nobel
laureate-microbiologist Joshua Lederberg to signify the microbial life in symbiosis
(under normal healthy conditions) with the human body (Prescott, 2017). In this con-
text, a fair definition of the microbiome could be: “the full complement of microbes
(bacteria, viruses, fungi, and protozoa), their genes, and genomes in or on the human
body”. Microbiota on the other hand, which is used similar to the term microbi-
ome, refers to an “ecological community of commensal, symbiotic and pathogenic
microorganisms found in all multicellular organisms studied to date from plants and
animals” (Prescott, 2017; Institute of Medicine, 2013). Hence, the difference between
microbiota and microbiome is that the latter could be more encompassing in the
sense that it refers not only to the organisms but also to the genome present and the
relevant implications.
The notion that something existing in the gut could produce health effects is indeed
ancient. Furthermore, the idea that fecal matter contains elements that could alter
favorable health outcomes to the recipients was also entertained since the antiquity.
The history of fecal microbial transplant (FMT) constitutes a characteristic example:
Although it is often considered a “novel” therapeutic strategy (Opritu, 2016), it was
being practiced while well-documented, certainly in terms of oral administration, in
various European medical crafts ever since ancient Greece. The practice is described
in numerous key works of medicine of 16th- and 17th-century Europe (Moore, 2019;
Faming et al., 2012), while further it was practiced in China since the Don-jin dynasty
(4th century AD) (Faming et al., 2012). FMT rectal delivery was used by American
doctor I.O. Wilson in 1910, particularly in the effort to instigate favorable changes in
fecal bacterial composition in patients with functional bowel disorders ameliorating
symptoms (Woodruff, 1910). Therefore, FMT cannot be considered novel, at least not
conceptually, whereas its historical and intercultural ubiquity may in all actuality pro-
vide solid support to an emerging scientific model, that of gut microbiome functioning
as a human body’s essential organ, itself comprised of several organisms which have
coevolved with human cells such that they are to some extent “us” and we are “them”.
On that basis, Alexander Khoruts, a pioneering researcher studying FMT, made
the argument that FTM-Clostridium difficile therapy must be viewed as a “trans-
plant” as opposed to a “drug” (Steve and Khoruts, 2014). Evidence regarding com-
mensal and symbiotic intestinal microbes continuously accumulates and supports
such an interpretation (Gray, 2017). Moreover, consistent with accepting the micro-
bial origin of human mitochondria (Gray, 2017), this organelle is another example
of coevolution through symbiosis where identifiable structures and unique DNA
(mtDNA) are evident (Moore, 2019).
Similarly, to several examples in biological sciences, the “microbiome” has
enjoyed significant biohype culminating to what some term a “microbiome zeitgeist”
Microbiome and Nutrition 73
(Prescott, 2017) leading to a “World microbiome day: June 27th”. Even though it is
well established and widely known both in the scientific community and in the public
that bacteria reside at various locales of the human body especially in the large intes-
tine (colon), the importance of the microbiome has gained significant attention due
to a significant body of scientific literature over the past 10–15 years indicating and
demonstrating that the size and type of microflora populations in the gut could well
influence health outcomes, spanning from immune responses to metabolic syndrome
(syndrome X), type 2 diabetes mellitus (T2DM), nonalcoholic steatohepatitis, cardio-
vascular disease (CVD) and some forms of cancer, particularly of colon (Bashiardes
et al., 2018). Thus, the microbiome is deemed integral to human physiology, health
and disease, while it arguably constitutes the most intimate connection that humans
extend to their external environment, mostly through diet. We typically consider the
large intestine (colon) as a site hosting a variety of populations of bacteria that reside
in a symbiotic mode at that habitat with the human body. While this is not the only
bodily location where bacteria reside, it appears to be a location with probably the
highest numbers and diversity of bacterial populations extending a rich variety of
metabolic and physiological influences.
The human microbiome, especially that of the gut, is being regarded an “essential
organ” (Bashiardes et al., 2018), with roughly 150 times more genes than the whole
human genome, while the human body contains at least 1,000 different species of
known bacteria. Microbiotic composition and function differ according to variations
in location, age, sex, race, and diet of the host (Steve and Khoruts, 2014). Although
determining precise numbers is rather challenging and of questionable accuracy, it
has been proposed that the number of human cells while at the order of 40 trillion
human cells (4 × 1013) in a typical human body is less than that of bacterial 1014 –1015
(Sender et al., 2016a, 2016b; Savage, 1977). There are recent estimates updating
the total number of bacteria in the 70 kg “reference man” to be 3.8 × 1013, while for
human cells, same estimates identify the dominant role of the hematopoietic lineage
to the total count (90%) and revise past estimates to 3.0 × 1013 human cells (Sender et
al., 2016a). The same analysis also updates the widely cited 10:1 ratio, showing that
the number of bacteria in the body is actually of the same order as the number of
human cells (Sender et al., 2016a).
The Human Microbiome Project (HMP) was initiated by the National Institutes of
Health (NIH) in 2007, with the majority of funding ($153 million of the $173 million
as of 2013 alone) coming from the NIH Common Fund (Institute of Medicine, 2013).
The project was initiated aiming to characterize the genomic makeup of all microbes
inhabiting the human body. Nonetheless, gradually even more so a growing number
of scientists place increasing emphasis on the importance of studying beyond the
identification on the microbes present but also the function of those microbes. HMP
used sequencing to examine the microbes associated with the human body. HMP’s
major purpose has been to generate resources for the research community, focusing
on building a “healthy cohort” reference database of human–microbiome genome
sequences (known as metagenomic sequences), computational tools for complex
metagenomic sequences analyses and clinical protocols for human–microbiome
sampling (Institute of Medicine, 2013).
74 The Gut Microbiome: Bench to Table
are several factors implicated to extending higher or lower degrees of influence to gut
microbiota composition, including genetics, health status, mode of delivery at birth
and environmental exposures.
While typically the consensus was that microbes were thought as from dangerous to
undesirable at best, the communities both scientific and lay do appreciate the vari-
ous aspects that the microbiome entails along with the beneficial ones that are health
promoting. Furthermore, it is increasingly more appreciated that it is in fact more
accurate to consider microbial communities (oftentimes diverse and in a dynamic
state) as opposed to microbes as units per se.
The development of a highly complex gut ecosystem in the human host begins
at or possibly even prior to birth, while the intestinal microbiota early colonization
depends on various environmental exposures. The taxa colonizing the otherways
relatively “sterile” newborn intestine are largely determined by the mode of deliv-
ery. Newborn intestinal colonization in the case of vaginal birth initiates while the
neonate passes through the birth canal. Neonatal’s ingestion of microorganisms
induces the gut microbiome and initiates its development. Moreover, infant feed-
ing depending on whether it is breastfeeding or formula-feeding further informs the
Microbiome and Nutrition 77
manner in which the intestinal microbiota will develop both in terms of quality and
quantity and overall bacterial demography. The early species are placed in the neo-
natal intestines through maternal feeding. Breastfeeding providing maternal milk
carbohydrates naturally selected to feed mutualist microbes in the infant’s micro-
biota provides significant support in the microbial growth. Beyond its nutritive value
both fort the infant and its intestinal microbiome, breastfeeding is further credited
with reducing infant mortality and limiting the risk of chronic diseases later in life.
Breastfed infants exhibit a Bifidobacteria-dominated microbiome inducing a tolero-
genic immune response in the gut, hence reducing the risk of pathogen colonization,
infection and overall gut inflammation (Wasielewski et al., 2016).
Human milk oligosaccharides (HMOs), the second most abundant milk carbohy-
drate after lactose, provide 5%–6% of milk’s energy content. Gut Bifidobacteria’s
growth is primarily supported by this type of carbohydrates. Interestingly, since
HMOs cannot be digested by humans, it requires microbial fermentation to produce
utilizable energy fuel for the host (i.e., human) in the form of SCFAs. Moreover, since
breast milk’s sialylated oligosaccharides prevent pathogen adhesion to epithelial cells,
they contribute significantly to infection prevention. Additionally, breast milk’s lipid
fraction further contributes to milk’s antipathogen properties. Breast milk’s triglycer-
ides are converted to antimicrobial fatty acids and monoglycerides by specific lipases
produced by the infant, which extend antiviral, -bacterial and -protozoan pathogen
properties, creating a hostile environment unsupportive of harmful microbe growth
(Wasielewski et al., 2016). Finally, breast milk is significantly advantageous in the
sense that is significantly enriched by a variety of antibodies reflective of mother’s
immunological profile as informed both by innate and by adaptive immunity.
Host-provided glycans are characterized by structural similarities with HMOs
and similar antimicrobial properties, offering an alternative to breast milk support
route for desirable microbes. The gel-like mucosal structure coating the intestinal
epithelium consists of proteins and O-glycosylated carbohydrate chains. This muco-
sal structure feeds the specific commensal bacteria, which in turn produce SCFAs,
while the host can capture the energy from SCFAs, thus contributing toward the
energy balance for manufacturing the mucus structure. This mutualistic relation-
ship results in benefit for the host because microbes occupy the glycan-binding sites,
thereby preventing pathogen crossing of the mucus layer which would cause inflam-
mation and ultimately infection (Wasielewski et al., 2016).
The symbiotic relationships between microbiota and host stem as a result of a
dynamically balanced habitat of symbionts and pathobionts. Perturbations affect-
ing the afore-described delicate balance leading to disturbance in the composition
of microbial communities, termed dysbiosis, typically result in disfavorable interac-
tions between the microbiome and the host, leading to increased disease suscepti-
bility. Various studies have demonstrated associations between intestinal dysbiosis
and chronic low-grade inflammation and metabolic dysfunctions. Thus, metabolic
syndrome, obesity and diabetes may be negatively impacted from changes in the gut
microbiome (Hemarajata and Versalovic, 2013; Thakur et al., 2014).
The gut microbiome may be affected by dietary alterations, infections and expo-
sure to antibiotics. Throughout infancy and early childhood, the aforementioned fac-
tors can alter the microbial balance in the individual’s gut. The infant’s intestinal
78 The Gut Microbiome: Bench to Table
FIGURE 3.1 Conceptual scheme: microbiome – diet interactions can lead to metabolic sig-
naling, energetics and disease risk modulation. (Adapted from: Sikalidis and Maykish, 2020.)
High levels of plasma lipopolysaccharides (LPS) with ectopic hepatic lipid accu-
mulation increase gut permeability, and low-grade endotoxemia have all been
demonstrated in animals whereby obesity was induced via exposure to high-fat or
high-fructose diets. High-fructose diets, in particular, have been strongly related to
both hepatic and extra-hepatic insulin resistance as well as obesity-related metabolic
disturbances via mechanisms involving gut microbiome and downstream intestinal
Microbiome and Nutrition 81
permeability (Ochoa et al., 2015). While sugars in general constitute a major car-
bon source modulating growth and virulence in a broad spectrum of gut pathogen
and pathobionts, refined/simple carbohydrates support overgrowth for opportunistic
bacteria including Clostridium difficile and Clostridium perfringens by induction of
biliary output (Wasielewski et al., 2016; Albenberg and Wu, 2014). Taken together,
these observations imply that high-fat/high-refined sugars diets can introduce unde-
sirable ecological alterations in the gut environment while intensifying host response
such as inflammation and differentiated glucose metabolism (Albenberg and Wu,
2014). Furthermore, simple carbohydrate overconsumption could produce unfavor-
able effects at both central and peripheral levels, such as fluctuations in secretion of
satiety-regulating peptides as well as neuropeptides. Further negative impact includes
increase of gut permeability leading to low-grade inflammation and liver disease as
well as induced blood–brain barrier (BBB) permeability, in turn affecting appetite and
mood/predisposition toward food, via the reward/dopamine axis related mechanism
(Ochoa et al., 2015). While fiber consumption seems to protect against inflammation,
the accumulation of simple sugars in the colon induces an increase of the osmotic
load, as well as the fermentation rate by the colonic microbiome, subsequently poten-
tially promoting abdominal pain, discomfort and various intestinal dysfunctions.
While enterocytes in the intestine will typically produce fatty acids, utilizing both
glucose and fructose as substrates, compared to glucose, fructose may be proven
a deprived substrate for de novo lipogenesis in enterocytes, while fatty acid accu-
mulation could worsen intestinal function. Consequently, excessive sucrose intake
has been recognized as a primary cause of metabolic syndrome, the toxicity result-
ing from excess fructose rather than sucrose, as fructose extends the induction of
glycation phenomena targeting proteins and, physiologically, its metabolic substrate
rapidly flows into de novo lipogenesis, which in turn strongly promotes liver inflam-
mation (MacDonald, 2016). Additionally, the pro-oxidative and pro-inflammatory
effects of fructose can lead to induced gut permeability and endotoxemia both of
which conditions do exacerbate chronic inflammation in the gut.
High sugar intake might disturb intestine homeostasis in a variety of mechanisms
and ways. The consumption of sugar-rich diets can alter the intestinal mucosa archi-
tecture. A high-sugar diet promotes intestinal stem cells differentiation into absorp-
tive enterocytes and secretive enteroendocrine cells in Drosophila, which causes the
gut to become thinner, and produces high levels of reactive oxygen species (Zhang et
al., 2017); specifically, high fructose intake can lead to reduced mucus thickness in
the colon and defensin secretion. Recent studies demonstrated that dietary fructose
worsens colitis in mice by altering the composition, localization and metabolism
of gut microbiota through mechanisms involving GLUT5 (glucose transporter 5)
expression (fructose transporter) (García-Montero et al., 2021).
Moreover, high glucose- and high fructose-fed mice have been shown to lose gut
microbial diversity (characterized by a reduced proportion of Bacteroidetes and a
markedly increased proportion of Proteobacteria) (Do et al., 2018). Dietary sugars
that are not absorbed at the intestinal level of the host and reach the colonic environ-
ment exposed to the microbiota also regulate gut colonization by beneficial microbes.
High-sucrose diets (when comprising 70% of the kilocalories from carbohydrates,
mainly in the form of sucrose) have previously been shown to elevate Clostridiales
82 The Gut Microbiome: Bench to Table
Unrefined grains, starchy and nonstarchy vegetables, legumes, and fruit constitute
dietary sources of a broad variety of nondigestible polysaccharides. Fiber, a collective
term for nondigestible polysaccharides divided into soluble and insoluble fibers, is not
easily digested or absorbed in the small intestine, but rather typically metabolized in
the distal colon. Cellulose is degraded by Bacteroides species, primarily associated
with plant polysaccharide degradation, or Ruminococcus, and degradation results
Microbiome and Nutrition 83
Intestinal bacteria produce a variety of fatty acids which in turn can extend health-
promoting effects. Bifidobacteria in the gut generate conjugated linoleic acid, which
is shown to influence fatty acid composition in the liver and adipose tissue in mouse
models (Devaraj et al., 2013; O’Shea et al., 2012). Additionally, as discussed earlier,
intestinal bacteria produce SCFAs such as acetate, butyrate, propionate via dietary
carbohydrate fermentation nondigestible by humans. Interestingly, de novo gluco-
neogenesis or lipid synthesis can be derived from certain SCFAs including propio-
nate, hence serving as a means for fuel production and energy yielding for the host
(human). Another interesting aspect of SCFAs is that they can extend signaling
properties and, in this sense, constitute the mode by which microbes can produce
metabolic signals affecting the metabolic regulation at the host level. In fact, SCFAs
can modulate carbohydrate metabolism as well as gut physiology via the stimulation
of mammalian peptide secretion while serving as fuel for gut epithelial cells. More
specifically, SCFAs can stimulate glucagon-like peptide 1 (GLP-1) secretion via the
G-protein-coupled receptor FFAR2 (free fatty acid receptor 2) in colon (Devaraj et
al., 2013). Through the stimulation of GLP-1 secretion, bacterially produced SCFAs
stipulate signals that downregulate glucagon secretion and stimulate glucose-depen-
dent insulin secretion, while promoting glucose homeostasis.
Peptide YY, a hormone released by ileal and colonic epithelial cells in response
to feeding seemingly able to suppress appetite, is also shown to be stimulated by
SCFAs (Zhou et al., 2006). Interestingly, butyrate-supplemented high-fat diets were
demonstrated to partially improve insulin resistance in diet-induced obese mice.
Simultaneously, diets low in specific types of carbohydrates are shown to yield
reduced butyrate-producing bacteria and fecal butyrate concentrations (Gao et al.,
2009), thus lending support to the notion that the type of fuel can determine bac-
terial metabolite output and by extension ensuing signaling. Propionate promotes
G-protein-coupled receptor 41 (GPR41)-mediated activation of sympathetic neurons,
in contrast to ketone bodies, thereby affecting energy homeostasis (Kimura et al.,
Microbiome and Nutrition 85
2011). This documented potential for sympathetic outflow modulation constitutes yet
another mechanism outlining the axis of gut microbiome–enteric nervous system–
energetics–metabolic homeostasis.
Interestingly, Roux-en-Y gastric bypass (RYGB) surgery, a major bariatric inter-
vention to treat morbid obesity, is shown to have an impact on the gut microbiome.
Enhanced populations of Proteobacteria at the expense of those of Bacteroidetes were
detected post-RYGB surgery. Such microbial population shifts plausibly result in
alterations of metabolite profiles as well as relative preponderance for various fatty
acids, including SCFAs. In animal experiments, nonobese rats subjected to RYGB
exhibited reduced amounts of both Firmicutes and Bacteroidetes while exhibited sig-
nificantly increased amounts (52-fold higher concentrations) of Proteobacteria com-
pared to sham-operated counterparts (Bouillot et al., 2010). While obesity can be
viewed as a chronic mild inflammatory or at least a pro-inflammatory state, the abun-
dance of butyrate-producing Faecalibacterium prausnitzii was demonstrated to be
negatively associated with biomarkers of inflammation prior and post-RYGB surgery,
indicative of the fact that such bacterial species could be contributors to a healthy gut
(Furet et al., 2010). Therefore, GI tract surgeries could extend a significant impact on
gut microbial profile ensuing SCFA production and immunological responses.
Antibiotic administration even at the subtherapeutic level constitutes a de facto
intervention in the GI tract and can alter the microbiome characteristics and by exten-
sion metabolic function. In an in vivo study, researchers administered subtherapeutic
doses of antibiotics to mice observing increase in adiposity and levels of incretin GIP-
1. Furthermore, significantly altered taxonomy in the microbiome was seen (increased
Lachnospiraceae and Firmicutes at the expense of Bacteroidetes). Moreover, changes
in metabolism-related genes (carbohydrates to SCFAs) were observed. More specifi-
cally, increased levels of acetate, propionate and butyrate were measured as well as
changes in the regulation of hepatic lipid and cholesterol metabolism. Therefore,
alteration of gut microbiota by antibiotic administration can result in the modula-
tion of murine metabolic homeostasis. In this context, significant concerns are posed
regarding antibiotics in the food chain and how these indirect exposures of consumers
to antibiotics, especially chronically, may influence the human gut microbiome and by
extension metabolic function and disease predisposition (Cho et al., 2012).
Additional microbial by-products with important relevance in host metabolism
and health include secondary bile acids, which affect glucose homeostasis by activat-
ing receptors similar to those activated by the parent compounds (e.g., farnesoid X
receptor-α), and trimethylamine (TMA), derived by choline degradation (Cella et al.,
2021). While TMA conversion to trimethylamine N-oxide is involved in CVD and
choline deficiency is related to nonalcoholic fatty liver disease and altered glucose
metabolism, secondary bile acids have been shown to be carcinogenic, thus signifi-
cantly increasing risk for colonic tumorigenesis (Ocvirk and O’Keefe, 2021).
Saturated fatty acids were reported to enhance dysbiosis as well as intestinal inflam-
mation in IL-10-deficient mice by inducing excessive growth of the bile-resis-
tant Gram-negative bacterium Bilophila wadsworthia. In separate experiments,
86 The Gut Microbiome: Bench to Table
feeding C57BL/6J mice high-saturated fat diet significantly promoted the growth
of 3 types of sulfidogenic bacteria in the colonic mucosa. These are hydrogen sul-
fide-producing bacteria as a metabolic by-product that damages intestinal barri-
ers while causing endotoxemia. Providing C57BL/6J mice with a high-saturated fat
dietary regime downregulated expression of tight junction proteins, contributing
to induced intestinal permeability, endotoxemia as well as elevated levels of LPS-
binding protein. Additionally, to higher fecal and plasma endotoxin levels, mice fed
the aforementioned diet exhibited lower Bifidobacteria populations but increased
Enterobacteriaceae populations in fecal culture. Laugerette et al. (2012) demon-
strated a significantly increased intestinal E. coli population along with elevated
both plasma and adipose inflammation in animals fed palm oil-based diets com-
pared to counterparts fed diets with unsaturated fat. Collectively, these data suggest
that diets high in saturated fat may negatively modulate the profile and subsequently
the function of gut microbiota, contributing to increased inflammation in animal
models (Alcock and Lin, 2015).
Researchers showed that high-fat diets resulted in a significant population
reduction of dominant bacteria and groups such as Bifidobacteria, Eubacterium,
Bacteroides and rectal Clostridium coccoides, constituting gut microflora. Such
microflora alterations in turn increase the ratio of Gram-negative to Gram-positive
bacteria including an increase in plasma LPS levels. Increased adiposity, body
weight, hepatic ectopic lipid accumulation, insulin resistance and type 2 diabetes
mellitus were all also associated with the aforementioned intestinal microflora
alterations (Estadella, 2013).
LPSs cause endothelial activation by way of receptor complex consisting of
Toll-like receptor 4 (TLR4), CD14 and myeloid differentiation factor 2 (MD2).
Endothelial activation is characterized as a pro-inflammatory state during which
the expression of leukocyte adhesion molecules increases, vascular integrity is lost,
cytokine production increases and human leukocyte antigen production is upreg-
ulated (Kelleher and Sikalidis, 2021). When the GI is in an inflammatory state,
metabolic endotoxemia occurs and bacterial LPS circulation increases by two- to
threefold (Bailey and Holscher, 2018). Therefore, a diet high in saturated fat may
further encourage a state of inflammation by upregulating LPS transport, while a
diet high in fiber may decrease the proportion of Gram-negative bacteria and subse-
quently prevent endotoxemia, thus contributing to an overall healthier gut environ-
ment (Kelleher and Sikalidis, 2021).
The role of omega 3 (ω-3) and omega 6 (ω-6) polyunsaturated fatty acids (PUFAs),
particularly omega 3, in regulating microbial metabolism by supporting optimal gut
microbial demography more so during early life is of significant interest. The omega-
class PUFAs, biosynthetic derivatives of α-linoleic acid and linolenic acid (LA) are
found mostly in fatty fish and some plant oils (Sandhu et al., 2016).
The majority of beneficial anaerobic bacteria including Roseburia, Bifidobacteria
and Lactobacillus are widely found in the colon, where PUFA metabolism from
Microbiome and Nutrition 87
the SPep, SPro and casein groups were 2.49 ± 0.60, 2.98 ± 1.12 and 2.59 ± 0.74, respec-
tively. Interestingly, while the rats fed SPro and SPep exhibited similar gut microbi-
ome profiles, SPep significantly promoted Lactobacillus and Phascolarctobacterium
growth. Their results indicated that SPep significantly increased the diversity of the
gut microbiota and elevated the probiotic portion of the bacterial gut population. The
authors concluded that both SPro and SPep were able to modify the demography of
gut microbiota in rats, with the effect of SPep being more desirable from a health
standpoint as judged by F/B ratio (Li et al., 2021).
In another study, Butteiger et al. demonstrated that soy protein compared to
milk protein in a Western-diet regime increases gut microbial diversity and reduces
serum lipids in golden Syrian hamsters (Butteiger et al. 2016). More specifically,
32, 6- to 8-week-old male golden Syrian hamsters were fed a Western diet contain-
ing 22% (%wt) milk protein isolate (MPI) as the single protein source for 3 week
followed by 6 week of one of four diets containing either of the following [22%
protein (%wt)]: MPI, soy protein concentrate, partially hydrolyzed soy protein
isolate (SPI1) or intact soy protein isolate. Serum lipids, hepatic gene expression
and gut microbial populations were evaluated. Sequencing of the 16S ribosomal
RNA gene revealed greater microbial diversity in each soy-fed group than in the
MPI-fed group (P < 0.05). The authors concluded that dietary protein sources in
male golden Syrian hamsters fed a Western diet differ in terms of how they affect
the gut microbiota with soy protein producing a favorable gut microbiome profile
while also potentially promoting the reduction of lipogenesis through alterations
of the gut microbial community (Butteiger et al. 2016). These in vivo studies lend
support to the notion that plant-based/-derived protein may constitute a dietary
approach that could extend health benefits, in addition to other shown aspects such
as N-balance with lower risk for allergies, also relative to the gut microbiome,
while also promoting sustainability and an eco-friendly approach to agriculture
and food production.
rearrangement, concluding that gut microbiome profile appears important for body
weight regulation (Ley et al., 2006). Interestingly, when gut microbiota of either
ob/ob mice or lean mice was transplanted to lean gnotobiotic mice, in mere 2 weeks,
mice that received microbiota from the ob/ob mice extracted more calories from
diet, while demonstrating significantly higher adiposity compared to mice receiving
the microbiota of lean mice. The results obtained sustenance a critical part of gut
microbiome regarding the pathogenesis of obesity and by extension obesity-related
disorders. In human experiments, Ley and colleagues (2006) as well as Ravussin
and colleagues (2011) successively examined fecal gut microbiota of 12 obese par-
ticipants who followed an annual weight loss program by engaging in a fat-restricted
or a carbohydrate-restricted, but always low-calorie diet. Analogous to similar in
vivo experiments, abundance of gut bacteria of the Bacteroidetes and Firmicutes
phyla was detected, while the microbiome exhibited noteworthy intraindividual
consistency over time. Prior to the low-calorie diet initiation, increased populations
of Firmicutes at the expense of those of Bacteroidetes were observed in the obese
individuals compared to nonobese controls. After weight loss, higher populations of
Bacteroidetes (3%–15%) with decreased populations of Firmicutes were seen. More
interestingly, the changes observed correlated quantitatively with weight loss rather
than dietary caloric content changes in terms of percentages.
In terms of definite dietary schemes, a recent study demonstrated that the
Mediterranean diet can positively affect resting metabolic rate as well as salivary
microbiota in humans subjects (Daniele et al., 2021). More specifically, salivary
microbiota composition and metabolic profile were analyzed in participants with
vegan (VEG) or Mediterranean (MED) long-term dietary patterns. The MED par-
ticipants demonstrated significantly higher percentages of Subflava and Prevotella
bacterial species as compared to VEG participants. Moreover, the authors reported
that MED participants showed higher basal metabolic rate (BMR) and lower respi-
ratory quotient (RQ). Furthermore, Prevotella abundance was demonstrated to be
inversely correlated with RQ and carbohydrate consumption (which was seen to
be lower in the MED participants), whereas Subflava percentages were demon-
strated to be positively correlated to BMR. Lactobacillus abundance, which was
inversely related to Subflava presence in MED participants, was associated with
decreased BMR (Harris-Benedict) values. Taken together, these observations
demonstrate positive effects of the Mediterranean diet on BMR and on the abun-
dance of microbial species associated with a better macronutrient metabolism
(Daniele et al., 2021).
The Mediterranean diet is becoming increasingly more popular due to growing
evidence regarding the role it can play in chronic disease prevention and immune
response modulation. Characterized by a diet rich in fruits, vegetables, fish, olive
oil, red wine and refined cereal products, the foods that are part of Mediterranean
diet are typically rich in fiber, omega-3 fatty acids, polyphenols, SCFAs, PUFAs,
phytosterols and antioxidants. Together, these bioactive compounds collectively and
synergistically extend anti-inflammatory and antioxidative properties, which may
be needed in counteracting accumulation of TMA N-oxides and the endotoxin LPS
(Kelleher and Sikalidis, 2021).
92 The Gut Microbiome: Bench to Table
FUTURE PERSPECTIVES
Whereas the link between gut microbiome, food and health is becoming increas-
ingly clearer, researchers are struggling to successfully and effectively manipulate
the microbiome as a form of treatment. Even though we have gained more knowledge
and understanding than ever before as to how the microbiome influences chronic
disease, it remains elusive as to how to change a person’s microbiome in a particular
favorable direction. There appears to be a clear need to assess how delivery and/or
interactive systems in the gut’s interface can be employed to affect bacterial type
and population size in the gut (Kristo et al., 2015) promoting optimal microbiome
demography, including but not solely limited to dietary schemes (Sikalidis, 2019). It
is clearly important to consider nutrition/diet perspectives as related to implications
with clinical nutrition and medical nutrition therapy. Beyond the studied relationship
between the microbiome and the obesity, further, there is significant evidence to
suggest that there are strong associations between the demographics/quality of the
microbiome and risk toward chronic diseases such as T2DM and ensuing CVD, as
well as inflammation and related pathologies (Sikalidis and Maykish, 2020), extend-
ing through the gut–brain axis to potential roles in Alzheimer’s as well as other
neurodegenerative diseases (Shukla et al., 2021).
After the successful completion of the human genome project approximately
two decades ago, Relman and Falkow urged the scientific community to embark
on a “second human genome project”, a large-scale genomic survey of our endog-
enous microflora (Relman and Falkow, 2001). Since then, the field of microbiome
research has greatly expanded, leading to large-scale efforts such as the NIH, HMP
and the Earth Microbiome Project. Those efforts alongside a significant increase in
the interest and ensuing research of the microbiome generated a significant body of
literature aiming to shed light on this human–microbiome symbiotic relationship.
There is ample evidence that microbiomes are networked and interconnected primar-
ily by means of the flows of mobile genetic elements (MGEs) (Haraoui, 2022). The
diversity, demography and profile of those MGEs have significantly expanded over
the past decades responsive to continuously growing selective pressures primarily
due to several anthropogenic forces applied. A microbial ontology more attuned to
these phenomena considering the aforementioned process could potentially provide a
more effective theoretical framework toward studying the microbiome and revealing
potential ways to utilize its plasticity for the benefit of human health.
REFERENCES
Albenberg LG, Wu GD. Diet and the intestinal microbiome: associations, functions, and
implications for health and disease. Gastroenterology. 2014;146(6):1564–1572.
Alcock J, Lin HC. Fatty acids from diet and microbiota regulate energy metabolism.
F1000Research. 2015;4(F1000 Faculty Rev):738.
Arnone D, Chabot C, Heba AC, Kökten T, Caron B, Hansmannel F, Dreumont N,
Ananthakrishnan AN, Quilliot D, Peyrin-Biroulet L. Sugars and gastrointestinal health.
Clin Gastroenterol Hepatol. 2021. doi: 10.1016/j.cgh.2021.12.011.
Bailey MA, Holscher HD. Microbiome-mediated effects of the Mediterranean diet on inflam-
mation. Adv Nutr. 2018;9(3):193–206. doi: 10.1093/advances/nmy013.
Microbiome and Nutrition 93
Garrido D, Nwosu C, Ruiz-Moyano S, Aldredge D, German JB, Lebrilla CB, Mills DA. Endo-
beta-N-acetylglucosaminidases from infant-gut associated bifidobacteria release com-
plex N-glycans from human milk glycoproteins. Mol Cell Proteom. 2012;11:775–785.
Gilbert K, Malick M, Madingou N, Touchette C, Bourque-Riel V, Tomaro L, Rousseau G.
Metabolites derived from omega-3 polyunsaturated fatty acids are important for car-
dioprotection. Eur J Pharmacol. 2015;769:147–153. doi: 10.1016/j.ejphar.2015.11.010.
Gray MW. Lynn Margulis and the endosymbiont hypothesis: 50 years later. Mol Biol Cell.
2017;28(10):1285–1287.
Haraoui LP. Networked collective microbiomes and the rise of subcellular ‘units of life’.
Trends Microbiol. 2022;30(2):112–119. doi: 10.1016/j.tim.2021.09.011.
Hemarajata P, Versalovic J. Effects of probiotics on gut microbiota: mechanisms of intestinal
immunomodulation and neuromodulation. Therap Adv Gastroenterol. 2013;6(1):39–51.
Heras VL, Melgar S, MacSharry J, Gahan CGM. The influence of the western diet on micro-
biota and gastrointestinal immunity. Annu Rev Food Sci Technol. 2022; doi: 10.1146/
annurev-food-052720-011032.
Holmes E, Wilson ID, Nicholson JK. Metabolic phenotyping in health and disease. Cell.
2008; 134(5):714–717.
Hussein HM, Elyamany MF, Rashed LA, Sallam NA. Vitamin D mitigates diabetes-associated
metabolic and cognitive dysfunction by modulating gut microbiota and colonic canna-
binoid receptor 1. Eur J Pharm Sci. 2022;170:106105. doi: 10.1016/j.ejps.2021.106105.
Institute of Medicine 2013. The Human–Microbiome, Diet, and Health: Workshop Summary.
Washington, DC: The National Academies Press.
Jones RB, Alderete TL, Kim JS, Millstein J, Gilliland FD, Goran MI. High intake of dietary
fructose in overweight/obese teenagers associated with depletion of Eubacterium
and Streptococcus in gut microbiome. Gut Microbes. 2019;10(6):712–719. doi:
10.1080/19490976.2019.1592420.
Kable ME, Chin EL, Storms D, Lemay DG, Stephensen CB. Tree-based analysis of dietary
diversity captures associations between fiber intake and gut microbiota composition in
a healthy U.S. Adult Cohort. J Nutr. 2021;nxab430. doi: 10.1093/jn/nxab430.
Kelleher AH, Sikalidis AK. The Effects of Mediterranean Diet on the Human Gut Microbiota;
a Brief Discussion of Evidence in Humans. Gastroenterol Hepatol. 2021;5(1):16.
doi:10.21926/obm.hg.2101056.
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barri-
ers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders.
Front Cell Neurosci. 2015;9:392.
Keyes KT, Ye Y, Lin Y, Zhang C, Perez-Polo JR, Gjorstrup P, Birnbaum Y. Resolvin E1
protects the rat heart against reperfusion injury. Am J Physiol Heart Circ Physiol.
2010;299(1):H153–H164. doi: 10.1152/ajpheart.01057.2009.
Kimura I, Inoue D, Maeda T, Hara T, Ichimura A, Miyauchi S, et al. Short-chain fatty acids
and ketones directly regulate sympathetic nervous system via G protein-coupled recep-
tor 41 (GPR41). Proc Natl Acad Sci U S A. 2011;108:8030–8035.
Kristo AS, Tzanidaki G, Lygeros A, Sikalidis AK. Bile sequestration potential of an edible
mineral (clinoptilolite) under simulated digestion of a high-fat meal; an in vitro investi-
gation. Food Funct. 2015;6(12):3818–3827.
Laugerette F, Furet JP, Debard C, Daira P, Loizon E, Géloën A, et al. Oil composition of
high-fat diet affects metabolic inflammation differently in connection with endotoxin
receptors in mice. Am J Physiol Endocrinol Metab. 2012;302(3):E374–E386.
Ley RE, Turnbaugh PJ, Klein S, Gordon IJ. Microbial ecology: human gut microbes associ-
ated with obesity. Nature. 2006;444(7122):1022–1023.
Li W, Li H, Zhang Y, Zhang C, Zhang J, Liu X. Differences in the gut microbiota com-
position of rats fed with soybean protein and their derived peptides. J Food Sci.
2021;86(12):5452–5465. doi: 10.1111/1750-3841.15948.
Microbiome and Nutrition 95
MacDonald IA. A review of recent evidence relating to sugars, insulin resistance and diabe-
tes. Eur J Nutr. 2016;55(Suppl 2):17–23. doi: 10.1007/s00394-016-1340-8.
Magnusson KR, Hauck L, Jeffrey BM, Elias V, Humphrey A, Nath R, Perrone A, Bermudez
LE. Relationships between diet-related changes in the gut microbiome and cognitive
flexibility. Neuroscience. 2015;300:128–140. doi: 10.1016/j.neuroscience.2015.05.016.
Mondot S, Lepage P. The human gut microbiome and its dysfunctions through the meta-omics
prism. Ann N Y Acad Sci. 2016;1372(1):9–19.
Moore AM. Nineteenth-century psychiatry of coprophilia and psychosis. Microbial Ecol
Health Dis. 2019;30:1546267.
Nicholson JK, Holmes E, Wilson ID. Gut microorganisms, mammalian metabolism and per-
sonalized health care. Nat Rev Microbiol. 2005;3(5):431–438.
Nicholson JK, Elliott P, Holmes E. Metabolome-wide association study identifies multiple
biomarkers that discriminate North and South Chinese populations at differing risks of
cardiovascular disease: Intermap study. J Proteome Res. 2010;9(12):6647–6654.
Ochoa M, Lallès JP, Malbert CH, Val-Laillet D. Dietary sugars: their detection by the gut-
brain axis and their peripheral and central effects in health and diseases. Eur J Nutr.
2015;54(1):1–24.
Ocvirk S, O’Keefe SJD. Dietary fat, bile acid metabolism and colorectal cancer. Semin
Cancer Biol. 2021;73:347–355. doi: 10.1016/j.semcancer.2020.10.003.
Opritu R, Bratu M, Opritu B. Fecal transplantation – the new, inexpensive, safe and rap-
idly effective approach in the treatment of gastrointestinal tract disorders. J Med Life.
2016;9(2):160–162.
O’Shea EF, Cotter PD, Stanton C, Ross RP, Hill C. Production of bioactive substances by
intestinal bacteria as a basis for explaining probiotic mechanisms: bacteriocins and con-
jugated linoleic acid. Int J Food Microbiol. 2012;152:189–205.
Prescott SL. History of medicine: origin of the term microbiome and why it matters. Hum
Microbiome J. 2017;4:24–25.
Ravussin Y, Koren O, Spor A, LeDuc C, Gutman R, Stombaugh J, et al. Responses of gut
microbiota to diet composition and weight loss in lean and obese mice. Obesity.
2011;20:738–747.
Relman DA, Falkow S. The meaning and impact of the human genome sequence for micro-
biology. Trends Microbiol. 2001;9(5):206–208. doi: 10.1016/s0966-842x(01)02041-8.
Rondeau I, Picard S, Bah TM, Roy L, Godbout R, Rousseau G. Effects of different dietary
ω-6/3 polyunsaturated fatty acids ratios on infarct size and the limbic system after
myocardial infarction. Can J Physiol Pharmacol. 2011;89(3):169–176. doi: 10.1139/
Y11-007.
Rossella C, Laura F, Grazia MM, Raffaele B, Antonio T, Maria P, Francesco V, Giovanni
G. The crosstalk between gut microbiota, intestinal immunological niche and visceral
adipose tissue as a new model for the pathogenesis of metabolic and inflammatory dis-
eases: the paradigm of type 2 diabetes mellitus. Curr Med Chem. 2022. doi: 10.2174/0
929867329666220105121124.
Rousseau G. Microbiota, a new playground for the Omega-3 polyunsaturated fatty acids in
cardiovascular diseases. Marine Drugs. 2021;19(2):54. doi: 10.3390/md19020054.
Salonen A, de Vos WM. Impact of diet on human intestinal microbiota and health. Annu Rev
Food Sci Technol. 2014;5(1):239–262.
Sandhu KV, Sherwin E, Schellekens H, Stanton C, Dinan TG, Cryan JF. Feeding the microbi-
ota-gut-brain axis: diet, microbiome and neuropsychiatry. Transl Res. 2016;179:223–244.
Sarkar A, Lehto SM, Harty S, Dinan TG, Cryan JF, Burnet PWJ. Psychobiotics and the
Manipulation of bacteria–gut–brain signals. Trends Neurosci. 2016;39(11):763–781.
Savage D. Microbial ecology of the gastrointestinal tract. Annu Rev Microbiol. 1977;31:107–133.
Sender R, Fuchs S, Milo R. Revised estimates for the number of human and bacteria cells in
the body. PLoS Biol. 2016a;14(8):e1002533.
96 The Gut Microbiome: Bench to Table
Sender R, Fuchs S, Milo R. Are we really vastly outnumbered? Revisiting the ratio of bacte-
rial to host cells in humans. Cell. 2016b;164:337–340.
Shukla PK, Delotterie DF, Xiao J, Pierre JF, Rao R, McDonald MP, Khan MM. Alterations in
the gut-microbial-inflammasome-brain axis in a mouse model of Alzheimer’s disease.
Cells. 2021;10(4):779. doi: 10.3390/cells10040779.
Sikalidis AK. From food for survival to food for personalized optimal health. A histori-
cal perspective of how food and nutrition gave rise to nutrigenomics. J Am Coll Nutr.
2019;38(1):84–95.
Sikalidis AK, Maykish A. The gut microbiome and type 2 diabetes mellitus; discussing a
complex relationship. Biomedicines. 2020;8(1):8. doi: 10.3390/biomedicines8010008.
Simopoulos AP. The importance of the omega-6/omega-3 fatty acid ratio in cardiovascular
disease and other chronic diseases. Exp Biol Med (Maywood). 2008;233(6):674–688.
doi: 10.3181/0711-MR-311.
Sonnenburg ED, Zheng H, Joglekar P, Higginbottom SK, Firbank SJ, Bolam DN, Sonnenburg
JL. Specificity of polysaccharide use in intestinal bacteroides species determines diet-
induced microbiota alterations. Cell. 2010;141:1241–1252.
Speliotes EK, et al. Association analyses of 249,796 individuals reveal 18 new loci associated
with body mass index. Nat Genet. 2010;42(11):937–948.
Steve LB, Khoruts A. Fecal microbiota transplantation: an interview with Alexander Khoruts.
Global Adv Health Med. 2014;3(3):73–80.
Thakur AK, Shakya A, Mohammed G, Emerald M, Kumar V. Gut-microbiota and mental
health: current and future perspectives. Pharmacol Clin Toxicol. 2014;2(1):1–15.
Tran Quang T, Gosselin A-A, Bourque-Riel V, Gilbert K, Charron T, Rousseau G.
Effect of Resolvin D1 on experimental myocardial infarction. Exp Clin Cardiol.
2014;20:6704–6712.
Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-
associated gut microbiome with increased capacity for energy harvest. Nature.
2006;444(7122):1027–1031.
Wasielewski H, Alcock J, Aktipis A. Resource conflict and cooperation between human
host and gut microbiota: implications for nutrition and health. Ann N Y Acad Sci.
2016;1372(1):20–28.
Woodruff IO. Organotherapy. J Adv Ther. 1910;28:551.
Yang W, Yu T, Huang X, Bilotta AJ, Xu L, Lu Y, Sun J, Pan F, Zhou J, Zhang W, Yao S,
Maynard CL, Singh N, Dann SM, Liu Z, Cong Y. Intestinal microbiota-derived short-
chain fatty acids regulation of immune cell IL-22 production and gut immunity. Nat
Commun. 2020;11(1):4457. doi: 10.1038/s41467-020-18262-6.
Yao Y, Cai X, Fei W, Ye Y, Zhao M, Zheng C. The role of short-chain fatty acids in immu-
nity, inflammation and metabolism. Crit Rev Food Sci Nutr. 2022;62(1):1–12. doi:
10.1080/10408398.2020.1854675.
Yap IK, Brown IJ, Chan Q, Wijeyesekera A, Garcia-Perez I, Bictash M, et al. Metabolome
wide association study identifies multiple biomarkers that discriminate North and South
Chinese populations at differing risks of cardiovascular disease: Intermap study. J
Proteome Res. 2010;9(12):6647–6654.
Zhang X, Jin Q, Jin LH. High sugar diet disrupts gut homeostasis though JNK and STAT
pathways in drosophila. Biochem Biophys Res Commun. 2017;487(4):910–916. doi:
10.1016/j.bbrc.2017.04.156.
Zhou J, Hegsted M, McCutcheon KL, Keenan MJ, Xi X, Raggio AM, Martin RJ. Peptide YY
and proglucagon mRNA expression patterns and regulation in the gut. Obesity (Silver
Spring). 2006;14:683–689.
Zoetendal EG, Raes J, van den Bogert B, Arumugam M, Booijink CC, Troost FJ, et al.
The human small intestinal microbiota is driven by rapid uptake and conversion of
simple carbohydrates. ISME J. 2012;6:1415–1426.
4 Models for Researching
the Gut Microbiome
Alison Lacombe and Vivian C.H. Wu
United States Department of Agriculture
Aleksandra S. Kristo
California Polytechnic State University
CONTENTS
Introduction...............................................................................................................97
Computational Models.............................................................................................. 98
In Vitro Models........................................................................................................ 100
Fecal Batch Cultures.......................................................................................... 100
Cell Culture........................................................................................................ 100
Ex Vivo Models................................................................................................... 101
3D Cell Culture.................................................................................................. 102
Microfluidics Systems and the “gut chip”.......................................................... 102
Simulator of the Human Intestinal Microbial Ecosystem (SHIME)....................... 103
Animal Models for Studying Gut Microbiota......................................................... 105
Insect Models..................................................................................................... 105
Avian Models..................................................................................................... 107
Murine Models................................................................................................... 107
Humanized Gnotobiotic Mice............................................................................ 109
Porcine Models.................................................................................................. 110
Zebrafish Model................................................................................................. 110
Fecal Microbiome Translocation Models............................................................... 111
Conclusion.............................................................................................................. 113
References............................................................................................................... 113
INTRODUCTION
Models are used to study gastrointestinal health to determine the impact of a
treatment, critical time points for administration, and potential mechanisms that drive
efficacy. For many studies pertaining to probiotics and prebiotics, complex microbio-
logical and metabolomics assays serve as proxy for microbiota function (Mabwi et al.,
2021). A “snapshot” of the gut microbiota composition and function can provide valu-
able insights into what factors alter compositional and metabolic changes subsequent
to dietary interventions (Mabwi et al., 2021). However, investigating discreet time
points only offers limited information regarding the dynamics of compositional and
DOI: 10.1201/b22970-6 97
98 The Gut Microbiome: Bench to Table
COMPUTATIONAL MODELS
Mathematical models can determine the statistical relationship between dietary
interventions and gastrointestinal health; however, these experiments are subject to
the assumptions used to build the models (Lamichhane, Sen, Dickens, Orešič, &
Bertram, 2018). “Snapshots” of the gut metagenome or metabolome often fail to
provide sufficient information on periodic patterns, interdependencies, or temporal
variations present within the microbiome (Lamichhane et al., 2018). Longitudinal
analysis can elucidate the changes over time and offer better context to the health
impacts of dietary interventions; however, this approach presents its own unique set
of challenges (Mabwi et al., 2021). Longitudinal studies in human populations are
often complicated by limited time points, sample dropout, and uneven sampling fre-
quency (Mabwi et al., 2021). Therefore, other models, such as animals or cell culture,
are often employed for complex computational analysis.
Interactions in a microbial community are driven by several mechanisms includ-
ing competition for nutrients, direct interactions between community members, and
interactions with the host and attachment sites in the host environment (Mabwi et al.,
2021). This network of bacterial interactions can be altered by the host’s age, diet,
disease initiation, or other exogenous perturbations (Mabwi et al., 2021). The combi-
nations of the correct models and computational tools are needed to infer ecological
relationships of microbiota and its associated metabolic functionality.
Temporal networks that depict how a connected system evolves with time have
emerged as useful tools for analyzing the gut microbiome. There are numerous uni-
variate and multivariate analytical methods that can test the significance of dietary
intervention at discrete time points or within a time series (Mabwi et al., 2021). These
methods have been applied to both metataxonomic and metabolomic temporal data.
For longitudinal studies where specific microbial features (e.g., taxa) are differen-
tially abundant, data smoothing, such as spline ANOVA, can discrete time points
into a polynomial curve (Mabwi et al., 2021). This analysis can estimate a difference
Models for Gut Microbiome 99
in function in the context of time with differential abundance. With the integration
of the difference function over a time interval, a statistical divergence in the data
set can be detected. Tools such as MetaDprof and MetaLonDA demonstrated the
specificity and sensitivity needed for detecting time periods of differentially abun-
dant features (Mabwi et al., 2021). Most notably, MetaLonDA can handle uneven
sample sizes and time intervals, which are common in human subject studies. For
example, using inputs from the DIABIMMUNE project, MetaLonDA found signifi-
cant differences between the Finnish and Russian cohorts in the relative abundances
of Bacteroides and Bifidobacterium species during the first year of life (Mabwi et
al., 2021). Additionally, MetaLonDA showed that Bacteroides is significantly more
abundant from day 96 to day 584 in Finnish infants, while Bifidobacterium is sig-
nificantly more abundant in Russian infants from day 96 to day 720 (Markowiak &
Ślizewska, 2017). This type of computational analysis can inform the treatment of
several gut-driven immune disorders often seen in children and provide valuable
regional population context.
Compared to taxonomic profiling and metagenomic data, there have been fewer
published works on metabolomic data. In dietary studies, the purpose of analyz-
ing metabolomic data is to determine whether two or more metabolite profiles are
significantly different. For a comparison of time series data from two experimental
groups, Hoteling-T2 statistic can be used as a measure of holistic differences (Chong
et al., 2019). When these studies employ multiple groups or levels of treatments,
multivariate empirical Bayes analysis of variance may be used to test for signifi-
cance (Chen et al., 2020). In cases where the specific metabolite might be unknown
or is overly complex to analyze, significant discriminatory biomarkers are used in
multivariate analysis (Park, Ufondu, Lee, & Jayaraman, 2020a). Linear mixed mod-
els offer an attractive option to investigate the longitudinal trend of metabolites and
identify metabolites that show significant concentration changes over time (Mabwi
et al., 2021). One promising technique is dynamic probabilistic principle coordi-
nate analysis, which models the correlations in multivariate data that occur due to
repeated measurements in time (Mabwi et al., 2021).
Integration of complex computation into microbiome studies is important. Even
with longitudinal studies, there is often a mismatch between microbial community
dynamics and sampling frequency. Most in vivo studies collect samples infrequently,
because of practical limitations, over days or weeks, whereas microbial community
dynamics in the gut microbiota can significantly vary within hours (Park, Ufondu,
Lee, & Jayaraman, 2020b). One possible solution is to use studies in parallel with
computational models to simulate the microbiome or metabolome dynamics to iden-
tify optimal sample collection (Park et al., 2020b). Computational and statistical
methods can rigorously address the challenges of microbiome temporal data analy-
sis. For mechanistic studies, a promising collection of software tools has become
available to support the analysis of large “omics” data sets from longitudinal studies
(Park et al., 2020b). There is no one size fits all regarding a standardized process for
reporting and comparing outcomes from different analyses. As a result, the choice
of a data analysis tool often relies on trial and error. In this regard, open-source plat-
forms that provide options to conduct multiple analyses on consistently formatted
and normalized data sets would be a powerful resource for elucidating diet.
100 The Gut Microbiome: Bench to Table
IN
In vitro models constitute an opportunity to study the gut microbiota in cases that an
in vivo study would be considered conceptually not feasible or unethical. In essence,
in vitro gut models function as chemostats, that is, bioreactors inoculated with fecal
microbiota and operated under physiological temperature, pH, and anaerobic condi-
tions (Ahmadi et al., 2019). Experimentation within these models is very flexible
with regards to studying multiple permutations/combinations of dietary ingredients
and their impact on gut microbial populations (Ahmadi et al., 2019). Ultimately,
cumulative in vitro studies can lead to robust in vivo approaches to investigating gut
microbiota functionality (Litten-Brown, Corson, & Clarke, 2010; Nguyen, Vieira-
Silva, Liston, & Raes, 2015; Ray & Dittel, 2015). In theory, each study yields com-
plementary results that strengthen the overall validity of each individual model and
distinguishes between the functionality of gut microbes and human processes.
Many of the microbes that reside within the gut are not cultivable using traditional
microbiological techniques such as agar Petri plates (Nagalingam, Kao, & Young,
2011). Preliminary studies of the human microbiome typically start with obtaining
donor specimens and culturing the microbes under anaerobic conditions in a biore-
actor (Ahmadi et al., 2019). The bioreactor setup can be as simple as an Erlenmeyer
flask inside a CO2 incubator on a stir plate or a complex system of pumps and valves
meant to closely simulate the physical system of the digestive tract (Ahmadi et al.,
2019). The complexity of the bioreactor setup is usually dictated by the question the
investigator is attempting to answer. For example, in determining if a potential food
ingredient is bifidogenic, investigators can quantify the presence of Bifidobacteria
using agar and simple biochemical tests. However, such setup cannot reveal the
ingredient’s effect on other important bacterial species or potential effects in human
physiology (Ahmadi et al., 2019; Vrieze et al., 2013a). For a comprehensive in vitro
or ex vivo analysis of microbiome shift with different dietary interventions, metage-
nomic analysis is still needed. For preliminary studies of the interactions of the
gut microbiome and specific nutrients, a video protocol can be found at www.jove.
com/v/59524/an-vitro-batch-culture-model-to-estimate-effects-interventional. This
protocol details a comprehensive and cost-effective way to use a bioreactor for study-
ing the compositional shift that occurs in a fecal sample following dietary amend-
ments (“An In Vitro Batch-culture Model to Estimate the Effects of Interventional
Regimens on Human Fecal Microbiota|Protocol,” n.d.).
In vitro human cell models are typically composed of polarized monolayers of single
or co-cultured carcinoma cells (Saygili, Dogan-Gurbuz, Yesil-Celiktas, & Draz, 2020).
These cell lines are distinguished based on their physiological properties and ability to
differentiate (Pearce et al., 2018). The intestinal epithelial consists of several cell types,
including enterocytes, goblet cells, stem cells, enteroendocrine cells, Tuft cells, M cells,
Models for Gut Microbiome 101
and Paneth cells, all of which can interact or sense the presence of the gut microbiota
and respond accordingly (Pearce et al., 2018). Caco-2 and mucus-secreting HT29-MTX
cell lines predominate the literature with regards to the study of probiotics and prebiot-
ics (Pearce et al., 2018). There are important considerations in selecting a cell culture
model, and decisions should be guided based on the research question asked.
Caco-2 cell line, derived from colorectal adenocarcinomas, is one of the oldest and
most utilized cell cultures in medical research (Pearce et al., 2018). This cell line can
function as an undifferentiated large intestinal cell or can spontaneously differenti-
ate into a cell closely resembling a small intestinal enterocyte (Ahmadi et al., 2012).
This ability to spontaneously differentiate is difficult to control and presents chal-
lenges in the reproduction of scientific studies, despite the fact that these Caco-2 cells
are widely used to study the protective effects of probiotics and prebiotics (Pearce
et al., 2018). Additionally, Caco-2 cells are commonly used in immunological studies
since they can express important pattern recognition receptors, Toll-like receptors,
which can provide information about the relative abundance of inflammatory mark-
ers known to contribute to the disease process (Payne et al., 2012).
HT-29 is another commonly utilized, polarized, and undifferentiated human
colorectal adenocarcinoma cell line (Paul et al., 2018). Recently, several subpopula-
tions have been selected to exhibit more “enterocyte-like” features, but in general,
spontaneous differentiation is less common compared to Caco-2 (Payne et al., 2012).
HT-29 cell lines have been extensively studied in the field of nutrition and host–
microbiome interactions because they produce important inflammatory cytokines,
such as interleukins and TNF-α (De Simone et al., 2015; Khan Mirzaei et al., 2016).
In addition, HT-29 provides an important benefit compared to Caco-2 in that HT-29
cells contain mucus-producing goblet cells. The ability to produce mucus is critical
for predicting immune function in the intestinal brush border and studying bacte-
rial–host interactions (Raja et al., 2012; Altamimi et al., 2016).
Prebiotic oligosaccharides have been shown to positively modulate both Caco-2
and HT-29 cells, and exhibit reduced inflammatory activity (Bode, 2009; Kelly,
2008). For example, the HT-29-MTX cell line, treated with methotrexate to produce
more mucin, has been a useful tool to show that oligosaccharides and a functional
mucin layer can reduce bacterial adhesion to the epithelium (Altamimi et al., 2016).
Studies with HT-29-MTX demonstrated that prebiotics can reduce the possibility of
an infection by reducing pathogens’ adhesion, migration, and internalization, includ-
ing Salmonella and E. coli. Co-cultures of Caco-2 and HT-29 render cell culture mod-
els more physiologically and functionally relevant, by combing the benefits of mucin
secretion and differentiation (Altamimi et al., 2016). In co-cultures of HT-29 Caco-2,
anti-adhesive effects of Streptococcus thermophilus and Lactobacillus acidophilus
against enteroinvasive E. coli were observed (Resta-Lenert and Barrett, 2003).
Ex ViVo models
Ex vivo models are models cultured outside of an organism, while closely mimic
functional live tissue systems and the complex cellular environments found in vivo
(Roeselers et al., 2013). These extended models are used to mimic the intestinal tract
in more detail by using advanced tissue engineering approaches. Ex vivo model
102 The Gut Microbiome: Bench to Table
systems can delve deeper into the complexity of the microbiome–host environment
and can probe the mechanism by which certain dietary components can improve
health outcomes (Roeselers et al., 2013).
3d cell culture
Modern 3D printing techniques allow for the creation of complex biological tissue
(Saygili et al., 2020). Bioprinting simultaneously combines living cells and biomateri-
als through a computer-aided design program to create 3D bioengineered living con-
structs that mimic natural tissue characteristics, including organs. This microscale
fabrication is commonly used for tissue engineering, regenerative medicine, micro-
biology, and the study of the gut microbiota (Saygili et al., 2020). In 3D bioprinting,
there are three major groups of techniques commonly used: layer-by-layer (stereo-
lithographic), line-by-line (extrusion-based), and droplet-based bioprinting (Saygili
et al., 2020). Extrusion-based bioprinting is the most common and affordable 3D
printing and creates complex, multilayered scaffolds and tissue constructs in bio-
medical applications. The stereolithography utilizes a photopolymerization process
in which UV light creates a pattern over a photopolymerizable liquid polymer, cross-
linking the light-sensitive polymers into a hardened layer (Patel, Lee, Park, Kim,
& Jeong, 2018). Droplet-based bioprinting utilizes multiple mechanisms to deposit
droplets of cell suspension in a high-throughput manner (Saygili et al., 2020). The
materials fed through the printer are typically hydrogels that have similar viscos-
elastic properties to that of biological material (Patel et al., 2018). Hydrogel-based
bio-ink materials typically have short-termed stability, the ability to retain nutrients,
and controlled cross-linking to facilitate bioprinter deposition (Patel et al., 2018).
The development of 3D tissue systems allowed for the modeling of host–bacte-
rial microbiome interactions in an organoid environment that closely resembles the
intestinal tract (Saygili et al., 2020). Organoid models for the gastrointestinal tract
were first developed in 2009 and have evolved into two distinct types, adult stem cell
(ASC) and pluripotent stem cell (PSC) derivatives (Saygili et al., 2020). ASC- derived
intestinal organoids are limited and consist only of basic epithelial cells such as pari-
etal cells, chief cells, and surface pit cells (Saygili et al., 2020). In comparison, PSC
has the capacity to differentiate into three germ layers (ectoderm, mesoderm, and
endoderm), mesenchymal cells, including smooth muscle cells, fibroblasts, and myo-
fibroblasts. However, to date, the cultivation of environmental cells, such as neural
cells, immune cells, and endothelial cells, is extremely difficult (Patel et al., 2018).
Without the environmental context of these other systems, organoids are prevented
from properly recapitulating the features of the actual organs designed to represent.
M
Recent advances in cell culture 3D printing and microfluidics have facilitated the
development of lab-on-a-chip systems that can be used to study the gut microbi-
ota. Gut chip and microfluidic systems typically house channels, chambers, sen-
sors, electrodes, and valves that allow for cellular growth media and air exchange
(Ashammakhi et al., 2020). Most microfluidic models of the gut involve an array of
Models for Gut Microbiome 103
microchannels that are separated by a porous and flexible membrane that allows for
nutrient exchange and elimination of waste, a design intended to simulate the bar-
rier between the intestinal lumen and the draining vasculature. The target cells are
seeded into channels and allowed to adhere, and then media is pumped through the
channels to simulate the microenvironment found in vivo (Ashammakhi et al., 2020).
Multiple cell types have been used within gut-on-a-chip devices depending on the
proposed research question. The most commonly used cell type is Caco-2 cells due
to their ease in culturing and capacity to differentiate in 3D cell culture, generating
an intestinal structure (von Martels et al., 2017). The presence of fluid flow to provide
biomimetic shear stress to cells is critical to this development, with the peristalsis-like
strain on cells inducing morphogenesis into 3D intestinal villi. However, the use of
Caco-2 if severely is limited by their lack of capacity to produce a significant mucosal
layer (von Martels et al., 2017). Therefore, co-culture with HT-29 cells and other types
of cells is an attractive option for many studies. In another gut model studying the
remodeling of extracellular matrix, Caco-2 cells were co-cultured with subepithelial
myofibroblasts to generate a full-thickness model of the intestine (Ashammakhi et al.,
2020). Co-cultures in 3D system can go beyond typical cell monolayers through the
formation of the villous structures, and the expression of tight junctions, presentation
of a brush layer, and production of mucous (von Martels et al., 2017).
The advent of gut chip technologies has allowed for the study of host–micro-
biota interaction, circumventing the use of costly animal models. A model micro-
fluidic environment consists of two compartments separated, one containing mixed
microbiota and the other enterocytes (Marzorati et al., 2014). To recreate physiologi-
cally relevant GIT conditions in order to study the gut microbiota, the following are
needed: (a) a mucosal area under shear stress for bacterial adherence; (b) transport
of low molecular weight metabolites; and (c) microaerophilic conditions. With this
model, it is possible to simulate bacterial adhesion and the indirect effect on cells.
Compared to traditional cell cultures, the constant nutrient cycling of microfluid-
ics systems allows for observations over longer periods with a lower risk of con-
tamination (Ashammakhi et al., 2020). Recent studies examined the intercellular
cross talk between Caco-2BBE cells and peripheral blood mononuclear cells in
response to challenges with dextran sulfate sodium (DSS), lipopolysaccharide (LPS)
endotoxin, probiotic VSL#3, and non-pathogenic E. coli (von Martels et al., 2017).
This system facilitated the isolation of each component of the signaling cascade to
identify the root cause of the inflammatory response. The gut epithelium disruption
with LPS activated the immune component of the model allowing for elucidation of
dysfunctional pathways, potentially not revealed by using complex animal models
(Ashammakhi et al., 2020).
aspects of digestion to the study design (Van De Wiele, Van Den Abbeele, Ossieur,
Possemiers, & Marzorati, 2015.). Simple fecal batch culture studies only utilize sin-
gle-stage chemostats to mimic colon conditions. These designs are often limited for
diet studies because environmental parameters such as pH, redox potential, available
nutrients, and microbial population dynamics constantly change throughout tran-
sit. To simulate in vivo conditions while still taking advantage of in vitro designs,
semicontinuous fermenters were developed where the intermittent supplementation
of nutritional medium (Van De Wiele et al., 2015). In contrast, fecal batch culture
studies that use one single fermenter ignore the heterogeneity in substrate availabil-
ity, fermentation activity, microbial composition, and other intrinsic characteristics.
The SHIME system is intended to mimic the microbiota, and the initial inoculum
originates from a donor’s fecal sample. Fecal samples capture a microbial com-
munity and metabolic shifts during transit from the proximal colon to the rectum.
It is important to consider that the fecal microbiome is significantly different from
the in vivo colon microbiome, both in terms of composition and metabolic activity
(Van De Wiele et al., n.d.). With that in mind, the colon is where the microbiome is
the most accessible sample for an individual intestinal community. The purpose of
the SHIME system is to adapt the fecal microbiome to the conditions present in the
different colon compartments (Van De Wiele et al., n.d.). The SHIME is typically
inoculated with fecal material sourced from individual donors, despite research due
to pool samples from several people. Pooling samples can partially account for the
interindividual variability that exists in microbiome composition and incorporates
properties from different enterotypes. Since there is enormous functional redun-
dancy of the gut microbiome, such pooled microbiome and the adaption process of
SHIME will shift the fermentation profile, not a strictly necessary step. However,
because SHIME models adapt the fecal microbiome to the simulated environment
of the colon, it often fails to capture function nuances. For example, the metabolism
of polyphenols, such as daidzein, isoxanthohumol, and catechins, is highly depen-
dent on an individual’s microbiome (van Duynhoven et al., 2011).
The mucosal microbiome is a crucial part of the gut microbial ecosystem because
of its proximity to host epithelial cells. It is thought to have an intrinsically higher
impact on human health because of the immunological activity that occurs (Van den
Abbeele et al. 2011). The mucosal microbiome is different from the luminal microbi-
ome in composition and, interestingly, the presence of important mucosal colonizers
such as Faecalibacterium prausnitzii (Willing et al. 2009). Access to the mucosal
environment in vivo is extremely difficult, and the development of gut simulators that
accurately mimic mucosal microbial colonization is needed. The SHIME can mimic
some of the mucosal microbial colonization by incorporation of mucin and other
compounds in which to establish a biofilm. One result of mucosal SHIME experi-
ments was the higher colonization rate of butyrate-producing Clostridium clusters IV
and XIVa (Van De Wiele et al., n.d.). This phylogenetic group is considered crucial
for delivering butyrate as a primary energy source to colonocytes and improves gut
barrier function by strengthening the tight junctions (Van De Wiele et al., n.d.).
This modular nature of SHIME is useful for diet studies and investigating the
microbiome’s ability to produce a bioactive metabolite (Possemiers et al. 2006). The
microbiome behavior in response to dietary inputs can be investigated by comparing
Models for Gut Microbiome 105
upper digestive tract to the colon. The residence times of the different gastric compart-
ments, the composition of the gastric juices, region-specific pH values, feed, feeding
regimes, and body temperatures are adapted in the SHIME setup leading to a simula-
tion of the targeted human or animal host. Finally, other features of the SHIME include
the gradual emptying of the gastric digest into the intestine compartment. The option of
running a dynamic pH profile in the gastric compartment enables the running experi-
ments with real food matrices or food constituents that need to undergo predigestion
and removal of sugar monomers or amino acids and peptides before the digest is trans-
ferred advancing to the colon compartment (Van De Wiele et al., n.d.).
Insects are the most diverse class of animals regarding species numbers and biomass
(Muñoz-Benavent, Pérez-Cobas, García-Ferris, Moya, & Latorre, 2021). The micro-
biota of insects, especially the gut microbiota, is as complex and rich as the phylogeny
and ecology of insects. Most insect species have endosymbiotic relationships with a spe-
cialized gut microbiota that contributes to health status and important nutritional roles
(Muñoz-Benavent et al., 2021). The insect intestine is inhabited by microorganisms
from all domains, including bacteria, fungi, archaea, protozoa, and viruses (Muñoz-
Benavent et al., 2021). As a result of their co-evolutionary histories with the host, these
microorganisms play essential roles in host physiology. Insect symbiotic systems are
either ectosymbiotic when the symbiont is on the surface of the host or endosymbiotic
when it lives inside specialized host cells (Muñoz-Benavent et al., 2021). For example,
termites gut endosymbiotic microbiota enables them to feed on a wood diet due to its
unique enzymatic capabilities. Cockroaches possess bacteriocytes, specialized cells
in the fat body of the insect, and an abundant and varied intestinal microbiota, whose
function is not fully understood (Muñoz-Benavent et al., 2021).
The combination of microorganisms populating the insect gut is driven by
several evolutionary and ecological factors, including phylogeny, diet, life stage,
and host environment (Muñoz-Benavent et al., 2021). Proteobacteria, Firmicutes,
Bacteroidetes, Actinobacteria, and Tenericutes are the most common phyla insect
guts, and the distribution of these phyla varies greatly among insect species. Diet is
a major driver of the gut microbiota composition in animals. Gene sequencing has
revealed the influence of diet on the gut microbiota of insects (Muñoz-Benavent
et al., 2021). With termites, there are major distinctions in the gut microbiota
106 The Gut Microbiome: Bench to Table
Birds represent a vertebrate class that plays a major role in natural ecosystems and
is with diverse gut microbiomes (Hird, Grond, Poulsen, & Jønsson, 2021). As with
most vertebrate species, the microbiome of birds is influenced to varying degrees by
host physiology and genetics, ecology and behavior, the environment, and diet (Hird
et al., 2021). Avian microbiomes are extremely diverse, and there is little similarity
between gut microbial community and host phylogeny. Low levels of host specificity
and high variability of the gut microbiome may result from the effects of a highly
diverse diet. Functional insights into avian gut bacteria stem mainly from poultry
studies, where gut microbes aid digestion, synthesize essential molecules for the host
(Hird et al., 2021), and interact with the immune system during microbiome estab-
lishment and development (Hird et al., 2021). However, poultry are unlikely to be
representative of all bird species. Nevertheless, the application of insights and meth-
ods developed for poultry may have implications for human diet and disease studies.
Birds represent important reservoirs and carriers of both animal and human
pathogens as well as antimicrobial resistance genes (Tellez et al., 2001). Bird micro-
biomes can reflect active infection status, and noninvasive methods exist to collect
fecal samples for migration and population-monitoring programs (Tellez et al., 2001).
Avian social behaviors impact the dynamics and assembly of gut bacterial communi-
ties (Tellez et al., 2001). Studies on social contact during breeding have yielded some
insights into the effects of family interactions on microbiomes. Group living may
also increase pathogen transfer among individuals who may select for diverse and
stable microbiomes that are more resilient to pathogen invasion, and such advantages
may be greater in social than in solitary species (Tellez et al., 2001).
Apart from chickens, few studies have investigated the cross talk between avian
immune systems and gut microbes, including gut symbiont responses to microbial
infections. In wild ducks, gut microbiome composition is significantly correlated with
influenza virus infections (Navarro-gonzalez et al., 2020), and in ostriches, coloniza-
tion of harmful bacteria can lead to dysbiosis and even death (Navarro-gonzalez et al.,
2020). Environmental microbial diversity can also impact both immune functions and
cloacal microbiomes of hosts (Zimmer-Faust, Steele, Griffith, & Schiff, 2020). Such
pattern-based and experimental manipulation of infections provides an initial foun-
dation to identify defensive microbial symbionts (Zimmer-Faust et al., 2020). These
insights can then be used to establish more precisely how microbes combat pathogens.
Rodents, mice, in particular, have become the most broadly and commonly used
model of choice for most studies in this emerging field. While both healthy human
and murine gut microbiota are dominated by the same two phyla (i.e., Bacteroidetes
and Firmicutes), studies by Ley et al. have demonstrated that approximately 85% of
the bacterial genera found in mice are not observed in humans under normal healthy
conditions (Ley et al., 2006). While similarities between the murine and human con-
dition have been observed, studies with mice have indicated changes in abundance
of bacterial phylotypes. In mice bacteria such as Tenericutes and Enterobacteriaceae
108 The Gut Microbiome: Bench to Table
Humanized gnotobiotic mice are produced by the inoculation of a human gut micro-
biota sample in GF mice. This murine model constitutes a powerful tool for gut
microbiota studies capturing a large part of the human gut microbiota phylogenetic
composition (100% of phyla, 11/12 classes, and ~88% of genus-level taxa) (Nguyen
et al., 2015). This approach has been widely employed in a plethora of studies since
it allows perturbations in a “human-like system” and is widely regarded as the gold
standard for confirming associations and trying to prove causality in gut microbiota
research (Faith, McNulty, Rey, & Gordon, 2011; Goodman et al., 2011).
Nevertheless, it must be emphasized that host–microbe relationships in human-
ized mouse models do not necessarily reflect the entire relationship spectrum seen
110 The Gut Microbiome: Bench to Table
in humans since the gut microbiota transplanted into a host (mouse) has not co-
evolved with the recipient. It has been reported that certain resident bacterial taxa
in the human gut microbiota are absent in the humanized mouse gut microbiota
(Turnbaugh et al., 2009).
Despite their limitations, humanized mice constitute one of the very few methods
to assess causality in microbiota research, and therefore, further development and
improvement of this approach is important.
Porcine Models
The porcine gut microbiota is increasingly and more rigorously studied due to the
scale of porcine husbandry industry, but also due to the similarities in anatomy,
physiology, and immunology to the human gastrointestinal tract (Litten-Brown
et al., 2010). An elegant and in-depth study of swine gut microbiota composition in
the Yorkshire pig breed demonstrated that human and pig microbiota shared similar
diversity patterns, with the two dominant phyla being Bacteroidetes and Firmicutes
(Lamendella, Santo Domingo, Ghosh, Martinson, & Oerther, 2011). However, at the
genus level, the swine gut microbiota harbors more Spirochaetes and Prevotella than
the human gut microbiota (Lamendella et al., 2011).
Another porcine model promising for microbiota research is the miniature pig.
Generally, miniature pigs develop obesity when fed ad libitum and are thus used as an
obesity and metabolic syndrome model. Specifically, the gut microbiota composition
of two miniature pig breeds, Gottingen and Ossabaw, was investigated for responses
to obesity induction (Meier & Bode, 2013; Pedersen et al., 2013), demonstrating that
the major phyla of miniature pig gut microbiota are Firmicutes and Bacteroidetes.
Interestingly, the two miniature pig breeds responded differently to an obesity-
inducing diet: Ossabaw gut microbiota displayed more of the characteristics of a
“healthy” obese microbiota, while Gottingen gut microbiota demonstrated alterations
analogous to metabolic syndromes, such as those found in gut microbiota profiles of
type 2 diabetic mice.
Zebrafish Model
Zebrafish is an attractive model used extensively for gut microbiota research due to
its small size, high fecundity, and full annotation of its genome. Given that several
gut functions and immune genes are conserved between zebrafish and mammals,
the zebrafish is an interesting model organism to investigate fundamental processes
underlying intestinal inflammation and injury (Brugman, 2016). As revealed by
genomic profiling, the zebrafish gut microbiota shares six bacterial divisions with
mice and five with humans, although marked differences exist in the relative abun-
dance of these phyla (Zhao et al., 2017). The microbiota of laboratory-reared zebraf-
ish intestine is dominated by Proteobacteria, while Firmicutes and Bacteroidetes
dominate in mice and humans (Rawls et al., 2004). Despite differences in the compo-
sition of their microbiota, the responses of zebrafish and mammals to microbial colo-
nization are similar. As an example, microarray analysis comparing the digestive
tracts of GF versus conventionally reared zebrafish revealed differential expression
Models for Gut Microbiome 111
of over 200 genes, of which nearly one-third were conserved in mice (Rawls et al.,
2004). The majority of these genes can be linked to epithelial proliferation, nutrient
metabolism, and immune responses. In terms of utilization of the zebrafish model,
Aryas-Jayo and colleagues investigated the effects of a high-fat diet (HFD) over a
period of 25 days on intestinal microbiota and inflammation in zebrafish. The con-
sumption of the HFD resulted in microbial dysbiosis, characterized by an increase in
the relative abundance of the phylum Bacteroidetes (Aryas-Jayo et al., 2018).
CONCLUSION
To elucidate causal relationships between prebiotics, probiotics, and gastrointestinal
health, multiple investigative approaches are needed that combine appropriate mod-
els with sensitive methods for data analysis. Modern approaches include in vitro,
in vivo, and ex vivo models, along with metagenomic and metabolomic analysis of
the community composition, its functional repertoire, and the by-products produced.
This information is often used to determine the dominant set of taxa that govern the
dynamics of multiple metabolic functions at the community level. In addition, infor-
mation can be revealed about the dynamics of the microbiome and the correlation
between specific taxa’s abundance with a metabolic function. However, due to the
complexity of the human body, it is rare that causation can be unequivocally estab-
lished using only one model.
REFERENCES
Ahmadi, S., Wang, S., Nagpal, R., Mainali, R., Soleimanian-Zad, S., Kitzman, D., & Yadav,
H. (2019). An In Vitro Batch-culture model to estimate the effects of interventional
regimens on human fecal microbiota. Journal of Visualized Experiments: JoVE, 149,
59524. https://doi.org/10.3791/59524
Altamimi, M., Abdelhay, O., Rastall, R.A. (2016). Effect of oligosaccharides on the adhesion
of gut bacteria to human HT-29 cells, Anaerobe, 39, 136–142.
An In Vitro Batch-culture Model to Estimate the Effects of Interventional Regimens on Human
Fecal Microbiota | Protocol. (n.d.). Retrieved August 12, 2020, from https://www.jove.
com/v/59524/an-vitro-batch-culture-model-to-estimate-effects-interventional
Antushevich, H. (2020a). Fecal microbiota transplantation in disease therapy. Clinica Chimica
Acta, 503, 90–98. https://doi.org/10.1016/J.CCA.2019.12.010
Antushevich, H. (2020b). Fecal microbiota transplantation in disease therapy. Clinica Chimica
Acta, 503(October 2019), 90–98. https://doi.org/10.1016/j.cca.2019.12.010
Arumugam, M., Raes, J., Pelletier, E., Le Paslier, D., Yamada, T., Mende, D. R., … Bork, P. (2011).
Enterotypes of the human gut microbiome. Nature, 473(7346). https://doi.org/10.1038/
nature09944
Ashammakhi, N., Nasiri, R., de Barros, N. R., Tebon, P., Thakor, J., Goudie, M., …
Khademhosseni, A. (2020, October 1). Gut-on-a-chip: Current progress and future
opportunities. Biomaterials, 255. https://doi.org/10.1016/j.biomaterials.2020.120196
Bode, L. (2009). Human milk oligosaccharides: Prebiotics and beyond. Nutrition Reviews.
https://doi.org/10.1111/j.1753-4887.2009.00239.x
CDC. (2020). Coronavirus Disease 2019 (COVID-19) Meat and Poultry Processing Workers
and Employers Exposure Risk among Meat and Poultry Processing Workers. 2019, 1–9.
Chen, T. M., Rui, J., Wang, Q. P., Zhao, Z. Y., Cui, J. A., & Yin, L. (2020). A mathematical
model for simulating the phase-based transmissibility of a novel coronavirus. Infectious
Diseases of Poverty, 9(1), 1–8. https://doi.org/10.1186/s40249-020-00640-3
Chong, P. P., Chin, V. K., Looi, C. Y., Wong, W. F., Madhavan, P., & Yong, V. C. (2019). The
microbiome and irritable bowel syndrome – A review on the pathophysiology, current
research and future therapy. Frontiers in Microbiology, 10(Jun). https://doi.org/10.3389/
fmicb.2019.01136
De Simone, V., Franzè, E., Ronchetti, G., Colantoni, A., Fantini, M.C., Di Fusco, D., Sica, G.S.,
Sileri, P., MacDonald, T.T., Pallone, F., Monteleone, G., & Stolfi, C. (2015). Th17-type
cytokines, IL-6 and TNF-α synergistically activate STAT3 and NF-kB to promote
colorectal cancer cell growth. Oncogene, 34, 3493–3503.
114 The Gut Microbiome: Bench to Table
Douglas, A. E. (2018). Drosophila and its gut microbes: A model for drug-microbiome inter-
actions. Drug Discovery Today: Disease Models, 28, 43–49. https://doi.org/10.1016/j.
ddmod.2019.08.004
Faith, J. J., McNulty, N. P., Rey, F. E., & Gordon, J. I. (2011). Predicting a human gut micro-
biota’s response to diet in gnotobiotic mice. Science, 333(6038). https://doi.org/10.1126/
science.1206025
Goodman, A. L., Kallstrom, G., Faith, J. J., Reyes, A., Moore, A., Dantas, G., & Gordon, J. I.
(2011). Extensive personal human gut microbiota culture collections characterized and
manipulated in gnotobiotic mice. Proceedings of the National Academy of Sciences of
the United States of America, 108(15). https://doi.org/10.1073/pnas.1102938108
Hird, S. M., Grond, K., Poulsen, M., & Jønsson, K. A. (2021). Avian gut microbiomes taking
flight, 1–13. https://doi.org/10.1016/j.tim.2021.07.003
Ianiro, G,. Maida, M., Burisch, J., Simonelli, C., Hold, G., Ventimiglia, M., Gasbarrini, A.,
& Cammarota, G. (2018). Efficacy of different faecal microbiota transplantation
protocols for Clostridium difficile infection: A systematic review and meta-analysis.
United European Gastroenterology Journal, 6(8), 1232–1244. https://doi.org/10.1177/
2050640618780762. PMID: 30288286; PMCID: PMC6169051.
Kelly, G. (2008). Inulin-type prebiotics – A review: Part 1. Alternative Medicine Review : A
Journal of Clinical Therapeutic, 13(4), 315–329.
Khan Mirzaei, M., Haileselassie, Y., Navis, M., Cooper, C., Sverremark-Ekström, E., &
Nilsson, A.S., (2016). Morphologically distinct Escherichia coli bacteriophages dif-
fer in their efficacy and ability to stimulate cytokine release In Vitro. Frontiers in
Microbiology, 7, 437.
Lamendella, R., Santo Domingo, J. W., Ghosh, S., Martinson, J., & Oerther, D. B. (2011).
Comparative fecal metagenomics unveils unique functional capacity of the swine gut.
BMC Microbiology, 11. https://doi.org/10.1186/1471-2180-11-103
Lamichhane, S., Sen, P., Dickens, A. M., Orešič, M., & Bertram, H. C. (2018). Gut metab-
olome meets microbiome: A methodological perspective to understand the relation-
ship between host and microbe. Methods, 149(March), 3–12. https://doi.org/10.1016/j.
ymeth.2018.04.029
Le Chatelier, E., Nielsen, T., Qin, J., Prifti, E., Hildebrand, F., Falony, G., … Yamada, T.
(2013). Richness of human gut microbiome correlates with metabolic markers. Nature,
500(7464). https://doi.org/10.1038/nature12506
Ley, R. E., Turnbaugh, P. J., Klein, S., & Gordon, J. I. (2006). Microbial ecology: Human gut
microbes associated with obesity. Nature, 444(7122). https://doi.org/10.1038/4441022a
Litten-Brown, J. C., Corson, A. M., & Clarke, L. (2010). Porcine models for the metabolic
syndrome, digestive and bone disorders: A general overview. Animal, 4(6). https://doi.
org/10.1017/S1751731110000200
Mabwi, H. A., Kim, E., Song, D. G., Yoon, H. S., Pan, C. H., Komba, E. V. G., … Cha, K. H. (2021).
Synthetic gut microbiome: Advances and challenges. Computational and Structural
Biotechnology Journal, 19, 363–371. https://doi.org/10.1016/j.csbj.2020.12.029
Markowiak, P., & Ślizewska, K. (2017). Effects of probiotics, prebiotics, and synbiotics on
human health. Nutrients, 9(9). https://doi.org/10.3390/NU9091021
Meier, P. P., & Bode, L. (2013). Health, nutrition, and cost outcomes of human milk feed-
ings for very low birthweight infants. Advances in Nutrition. https://doi.org/10.3945/
an.113.004457
Molly, K., Vande Woestyne, M., & Verstraete, W. (1993). Development of a 5-step
multi-chamber reactor as a simulation of the human intestinal microbial ecosys-
tem. Applied Microbiology and Biotechnology. May; 39(2), 254–258. https://doi.
org/10.1007/BF00228615. PMID: 7763732
Models for Gut Microbiome 115
Marzorati, M., Vanhoecke, B., De Ryck, T. et al. (2014). The HMI™ module: a new tool to
study the Host-Microbiota Interaction in the human gastrointestinal tract in vitro. BMC
Microbiology, 14, 133. https://doi.org/10.1186/1471-2180-14-133
Muñoz-Benavent, M., Pérez-Cobas, A. E., García-Ferris, C., Moya, A., & Latorre, A.
(2021). Insects’ potential: Understanding the functional role of their gut microbiome.
Journal of Pharmaceutical and Biomedical Analysis, 194. https://doi.org/10.1016/j.
jpba.2020.113787
Nagalingam, N. A., Kao, J. Y., & Young, V. B. (2011). Microbial ecology of the murine gut asso-
ciated with the development of dextran sodium sulfate-induced colitis. Inflammatory
Bowel Diseases, 17(4). https://doi.org/10.1002/ibd.21462
Nguyen, T. L. A., Vieira-Silva, S., Liston, A., & Raes, J. (2015). How informative is the mouse
for human gut microbiota research? DMM Disease Models and Mechanisms, 8(1).
https://doi.org/10.1242/dmm.017400
Niederwerder, M. C. (2018). Fecal microbiota transplantation as a tool to treat and reduce
susceptibility to disease in animals. Veterinary Immunology and Immunopathology,
206(October), 65–72. https://doi.org/10.1016/j.vetimm.2018.11.002
Park, S. Y., Ufondu, A., Lee, K., & Jayaraman, A. (2020a). Emerging computational tools
and models for studying gut microbiota composition and function. Current Opinion in
Biotechnology, 66, 301–311. https://doi.org/10.1016/J.COPBIO.2020.10.005
Park, S. Y., Ufondu, A., Lee, K., & Jayaraman, A. (2020b). Emerging computational tools
and models for studying gut microbiota composition and function. Current Opinion in
Biotechnology, 66, 301–311. https://doi.org/10.1016/j.copbio.2020.10.005
Patel, M., Lee, H. J., Park, S., Kim, Y., & Jeong, B. (2018). Injectable thermogel for 3D culture of
stem cells. Biomaterials, 159, 91–107. https://doi.org/10.1016/j.biomaterials.2018.01.001
Paul, W., Marta, C., & Van De Wiele, T. (2018, August 1). Resolving host–microbe interac-
tions in the gut: The promise of in vitro models to complement in vivo research. Current
Opinion in Microbiology, 44, 28–33. https://doi.org/10.1016/j.mib.2018.07.001
Payne, A. N., Zihler, A., Chassard, C., & Lacroix, C. (2012, January). Advances and perspec-
tives in in vitro human gut fermentation modeling. Trends in Biotechnology, 30, 17–25.
https://doi.org/10.1016/j.tibtech.2011.06.011
Pearce, S. C., Coia, H. G., Karl, J. P., Pantoja-Feliciano, I. G., Zachos, N. C., & Racicot, K.
(2018, November 12). Intestinal in vitro and ex vivo models to study host-microbiome
interactions and acute stressors. Frontiers in Physiology, 9. https://doi.org/10.3389/
fphys.2018.01584
Pedersen, A. N., Kondrup, J., & Børsheim, E. (2013). Health effects of protein intake in healthy
adults: a systematic literature review. Food & Nutrition Research, 30, 57. https://doi.
org/10.3402/fnr.v57i0.21245. PMID: 23908602; PMCID: PMC3730112.
Peloquin, J. M., & Nguyen, D. D. (2013). The microbiota and inflammatory bowel dis-
ease: Insights from animal models. Anaerobe, 24. https://doi.org/10.1016/j.anaerobe.
2013.04.006
Possemiers, S., Bolca, S., Grootaert, C., Heyerick, A., Decroos, K., Dhooge, W., De Keukeleire D.,
Rabot, S., Verstraete, W., Van de Wiele, T. (2006). The prenylflavonoid isoxantho-
humol from hops (Humulus lupulus L.) is activated into the potent phytoestrogen
8-prenylnaringenin in vitro and in the human intestine. Journal of Nutrition, Jul;
136(7), 1862–1867. https://doi.org/10.1093/jn/136.7.1862. PMID: 16772450.
Raja, M., Puntheeranurak, T,. Hinterdorfer, P., Kinne, R. (2012). Chapter Two - SLC5 and
SLC2 Transporters in epithelia—cellular role and molecular mechanisms. Current
Topic in Membrane, 70, 29–76.
Rawls, J.F., Samuel, B.S., & Gordon, J.I. (2004). Gnotobiotic zebrafish reveal evolutionarily
conserved responses to the gut microbiota. PNAS, 101(13), 4596–4601.
116 The Gut Microbiome: Bench to Table
Ray, A., & Dittel, B. N. (2015). Interrelatedness between dysbiosis in the gut microbiota due
to immunodeficiency and disease penetrance of colitis. Immunology, 146(3). https://doi.
org/10.1111/imm.12511
Resta-Lenert, S., & Barrett, K.E. (2003). Live probiotics protect intestinal epithelial cells from
the effects of infection with enteroinvasive Escherichia coli (EIEC). Gut, 52(7), 988–
997. https://doi.org/10.1136/gut.52.7.988. PMID: 12801956; PMCID: PMC1773702.
Roeselers, G., Ponomarenko, M., Lukovac, S., & Wortelboer, H. M. (2013). Ex vivo sys-
tems to study host-microbiota interactions in the gastrointestinal tract. Best Practice
and Research: Clinical Gastroenterology, 27, 101–113. https://doi.org/10.1016/j.
bpg.2013.03.018
Saygili, E., Dogan-Gurbuz, A. A., Yesil-Celiktas, O., & Draz, M. S. (2020, June 1). 3D
bioprinting: A powerful tool to leverage tissue engineering and microbial systems.
Bioprinting, 18. https://doi.org/10.1016/j.bprint.2019.e00071
Settanni, C. R., Ianiro, G., Bibbò, S., Cammarota, G., & Gasbarrini, A. (2021a). Gut micro-
biota alteration and modulation in psychiatric disorders: Current evidence on fecal
microbiota transplantation. Progress in Neuro-Psychopharmacology and Biological
Psychiatry, 109, 110258. https://doi.org/10.1016/J.PNPBP.2021.110258
Settanni, C. R., Ianiro, G., Bibbò, S., Cammarota, G., & Gasbarrini, A. (2021b). Gut micro-
biota alteration and modulation in psychiatric disorders: Current evidence on fecal
microbiota transplantation. Progress in Neuro-Psychopharmacology and Biological
Psychiatry, 109(August 2020). https://doi.org/10.1016/j.pnpbp.2021.110258
Tellez, G., Petrone, V. M., Escorcia, M., Morishita, T. Y., Cobb, C. W., & Villasenõr, L.
(2001). Evaluation of avian-specific probiotic and Salmonella enteritidis-, Salmonella
typhimurium-, and Salmonella Heidelberg-specific antibodies on cecal colonization
and organ invasion of Salmonella enteritidis in broilers. Journal of Food Protection,
64(3), 287–291. https://doi.org/10.4315/0362-028X-64.3.287
Turnbaugh, P. J., Ridaura, V. K., Faith, J. J., Rey, F. E., Knight, R., & Gordon, J. I. (2009).
The effect of diet on the human gut microbiome: A metagenomic analysis in human-
ized gnotobiotic mice. Science Translational Medicine, 1(6). https://doi.org/10.1126/
scitranslmed.3000322
Van de Wiele, T., Van den Abbeele, P., Ossieur, W., Possemiers, S., Marzorati, M. (2015). The
Simulator of the Human Intestinal Microbial Ecosystem (SHIME®). In The Impact of
Food Bioactives on Health, Verhoeckx, K., Cotter, P., López-Expósito, I., Kleiveland,
C., Lea, T., Mackie, A., Requena, T., Swiatecka, D., & Wichers, H. (Eds.). Springer,
Cham. https://doi.org/10.1007/978-3-319-16104-4_27
Van den Abbeele, P., Roos, S., Eeckhaut, V., MacKenzie, D.A., Derde, M., Verstraete, W.,
Marzorati, M., Possemiers, S., Vanhoecke, B., Van Immerseel, F., Van de Wiele,
T. (2012). Incorporating a mucosal environment in a dynamic gut model results in
a more representative colonization by lactobacilli. Microbial Biotechnology, 106–
115. https://doi.org/10.1111/j.1751-7915.2011.00308.x. PMID: 21989255; PMCID:
PMC3815277.
van Duynhoven, J., Vaughan, E.E., Jacobs, D.M., Kemperman, R.A., van Velzen, E.J., Gross,
G., Roger, L.C., Possemiers, S., Smilde, A.K., Doré, J., Westerhuis, J.A., Van de Wiele,
T. (2011). Metabolic fate of polyphenols in the human superorganism. Proceedings of
the National Academy of Sciences of the United States of America, 108 Suppl, 4531–
4538. https://doi.org/10.1073/pnas.1000098107. Epub 2010 Jun 25. PMID: 20615997;
PMCID: PMC3063601.
von Martels, J. Z. H., Sadaghian Sadabad, M., Bourgonje, A. R., Blokzijl, T., Dijkstra, G.,
Faber, K. N., & Harmsen, H. J. M. (2017, April 1). The role of gut microbiota in health and
disease: In vitro modeling of host-microbe interactions at the aerobe-anaerobe interphase
of the human gut. Anaerobe, 44, 3–12. https://doi.org/10.1016/j.anaerobe.2017.01.001
Models for Gut Microbiome 117
Vrieze, A., De Groot, P. F., Kootte, R. S., Knaapen, M., Van Nood, E., & Nieuwdorp, M.
(2013a). Fecal transplant: A safe and sustainable clinical therapy for restoring intes-
tinal microbial balance in human disease? Best Practice and Research: Clinical
Gastroenterology, 27, 127–137. https://doi.org/10.1016/j.bpg.2013.03.003
Vrieze, A., De Groot, P. F., Kootte, R. S., Knaapen, M., Van Nood, E., & Nieuwdorp, M.
(2013b). Fecal transplant: A safe and sustainable clinical therapy for restoring intes-
tinal microbial balance in human disease? Best Practice and Research: Clinical
Gastroenterology, 27(1), 127–137. https://doi.org/10.1016/j.bpg.2013.03.003
Vrieze, A., De Groot, P. F., Kootte, R. S., Knaapen, M., Van Nood, E., & Nieuwdorp, M.
(2013c). Fecal transplant: A safe and sustainable clinical therapy for restoring intes-
tinal microbial balance in human disease? Best Practice and Research: Clinical
Gastroenterology, 27(1), 127–137. https://doi.org/10.1016/j.bpg.2013.03.003
Wang, T., Wu, J., Qi, J., Hao, L., Yi, Y., & Zhang, Z. (2016). Kinetics of inactivation of
Bacillus subtilis subsp. niger spores and Staphylococcus albus on paper by chlorine
dioxide gas in an enclosed space. Applied and Environmental Microbiology. https://doi.
org/10.1128/AEM.03940-15
Willing, B., Halfvarson, J., Dicksved, J., Rosenquist, M., Järnerot, G., Engstrand, L., Tysk, C.,
Jansson, J.K. (2009). Twin studies reveal specific imbalances in the mucosa-associated
microbiota of patients with ileal Crohn's disease. Inflammatory Bowel Disease, 15(5),
653–660. https://doi.org/10.1002/ibd.20783. PMID: 19023901.
Wu, W. K., Chen, C. C., Panyod, S., Chen, R. A., Wu, M. S., Sheen, L. Y., & Chang, S. C.
(2019). Optimization of fecal sample processing for microbiome study—The journey
from bathroom to bench. Journal of the Formosan Medical Association, 118(2), 545–
555. https://doi.org/10.1016/J.JFMA.2018.02.005
Zhao, Y., Czilwik, G., Klein, V., Mitsakakis, K., Zengerleabc, R., & Paust, N. (2017). C-reactive
protein and interleukin 6 microfluidic immunoassays with on-chip pre-stored reagents
and centrifugo-pneumatic liquid control. Lab on a Chip, 17, 166.
Zimmer-Faust, A. G., Steele, J. A., Griffith, J. F., & Schiff, K. (2020). The challenges of micro-
bial source tracking at urban beaches for quantitative microbial risk assessment (QMRA).
Marine Pollution Bulletin, 160. https://doi.org/10.1016/J.MARPOLBUL.2020.111546
5 Dietary Modulation
of Gut Microbiota by
Cultured Products
Salam A. Ibrahim and Rabin Gyawali
North Carolina A&T State University
Raphael D. Ayivi
North Carolina A&T State University
University of North Carolina Greensboro
Hafize Fidan
University of Food Technologies
Saeed Paidari
Islamic Azad University
Reza V. Bakhshayesh
Agricultural Biotechnology Research Institute of Iran
University of Tabriz
CONTENTS
Introduction ............................................................................................................ 120
Biotherapeutic Properties of Cholesterol-Lowering Probiotics ............................. 121
Concept of Probiotics ........................................................................................ 121
Probiotic Microorganism................................................................................... 122
Important Criteria in Selecting Probiotics......................................................... 122
Synthesis and Metabolism of Plasma Lipoprotein ............................................ 124
Cholesterol-Lowering of Probiotics .................................................................. 125
Cholesterol-Lowering Effect Mechanisms........................................................ 126
Decongesting Bile Salts Using Bile Salt Hydrolyzing Enzyme........................ 126
Cholesterol Deposition with Conjugated Bile Salt ........................................... 128
Cholesterol Integration into Bacterial Cell Walls ......................................... 128
Lowering Serum Cholesterol by Probiotics ................................................. 129
Binding of Cholesterol ................................................................................. 130
Other Mechanisms........................................................................................ 131
Relationship between Nutrition and Gut Microbiota ................................... 132
INTRODUCTION
Proper diet and nutrition are fundamentally important for human health and meta-
bolic development and have been advocated by various stakeholders in the quest to
decrease the surging rate of health and metabolic diseases. More specifically, the
human gut microbiota has been associated with the proper regulation and functional-
ity of the human metabolic and immune system (Gentile & Weir, 2018). As a result of
the important role of the gut microbiota in combatting diseases and promoting health
and wellness, it is critical for the modulatory functionality of the gut microbiota to
be adequately supported in its functionality. Cultured food products, probiotics, and
fermented functional foods have all been proposed as conventional approaches to
naturally boosting the modulatory effect of the gut microbiota. Cultured food prod-
ucts allude to all fermented dairy or milk products or fermented food beverages with
lactic acid bacteria as a key culturing agent (Ayivi et al., 2020). Moreover, the grow-
ing demand by consumers and dietary health experts to adopt cleaner and greener
strategies in enhancing the human immune system warrants rapid attention and a
paradigm shift away from chemical supplements that could pose a human health
risk coupled with long-term side effects. The interplay between diet, nutrition, and
probiotic consumption results in a synergistic effect on the gut microbiota that boosts
the immune system in its fight against attacks from foreign bodies, pathogens, and
disease-causing organisms (Yang et al., 2020). Probiotics have been shown to pos-
sess a high therapeutic effect on human health. Moreover, studies have confirmed the
immunomodulatory properties of probiotics and diet on the gut microbiota through
the promotion of beneficial bacteria and the suppression of deleterious microor-
ganisms (Ibrahim et al., 2021). However, there is little available research that has
addressed the role of cultured products and diet together and their contingent effect
on the modulation of the gut microbiota.
This chapter thus embraces a nutraceutical concept and elucidates the promising
and potential role of diet, probiotics, and nutrition in the modulatory action of the gut
microbiota. We will also discuss the role of various factors such as cultured products,
probiotics, and fermented functional foods in gastrointestinal (GI) disease prevention,
Dietary Modulation of Gut Microbiota 121
BIOTHERAPEUTIC PROPERTIES OF
CHOLESTEROL-LOWERING PROBIOTICS
C
Probiotic microorganism
Lactobacillus and Bifidobacterium are considered as the most widely and commonly
used probiotic microorganisms nowadays (Nami et al., 2018). In addition, other
probiotic strains that have gained prominence and consumer acceptance include
Lactococcus (Shehata, El-Sahn, El Sohaimy, & Youssef, 2019), Streptococcus
(Shastri et al., 2020), Enterococcus (Nami et al., 2019), Bacillus (Lim, Oh, Yu, &
Kim, 2021), Saccharomyces (Rodríguez-Nogales et al., 2018), Propionibacterium
(Nair & Kollanoor Johny, 2018), Leuconostoc (Le & Yang, 2019), Weissella (Yadav,
Yadav, Singh, Singh, & Mani, 2019), or Escherichia coli originating from the GIT
(Behrouzi et al., 2020) or clostridia (Liu, Xie, Wan, & Deng, 2020) (Table 5.1).
Although pathogenic strains such as Bacillus anthracis and Bacillus cereus are nota-
ble in the Bacillus family, their toxicity is strain-dependent, and some strains do not
create enterotoxins, thus making them suitable for use as probiotics.
Probiotic applications in animal food and feed are also on the rise, but this must be
done with caution because the strain can spread from animal to human. Consequently,
stringent standards for probiotic safety and quality must be implemented and
TABLE 5.1
Some Known Lactic Acid and Probiotic Bacteria
Probiotic Microorganisms
Lactobacillus Bifidobacterium Other Lactic Acid
spp. spp. Bacteria Nonlactic Acid Bacteria
L. acidophilus B. adolescentis Enterococcus faecalis Bacillus cereus var. to yoi
B. coagulans GBI-30
L. casei B. animalis E. faecium E. coli strain nissle
L. crispatus B. bifidum Lactococcus lactis Propionibacterium freudenreichii
L. acidophilus B. breve Leuconostoc mesenteroides Saccharomyces cerevisiae
L. gallinarum B. infantis Pediococcus acidilactici S. boulardii
L. gasseri B. adolescentis Sporolactobacillus inulinus
L. johnsonii B. animalis Streptococcus thermophilus
L. paracasei B. bifidum Weissella paramesenteroides
L. plantarum B. breve
L. reuteri B. lactis
L. rhamnosus B. longum
L. brevis
L. fermentum
L. salivarius
L. helveticus
Dietary Modulation of Gut Microbiota 123
Apart from adhesion to mucus and/or human epithelial cells and cell lines, the prin-
cipal tests described above are identical to the Indian Council of Medical Research
and Department of Biotechnology (ICMR-DBT) criteria for probiotic strain in vitro
characterization (Ganguly et al., 2011). The Joint FAO/WHO Working Group advises
at a minimum the following screening tests in terms of organism safety (FAO &
WHO, 2002):
124 The Gut Microbiome: Bench to Table
The following assays for microbiological safety evaluation are recommended by the
ICMR-DBT guidelines (Ganguly et al., 2011):
Nonetheless, the significance of these metrics is still debatable due to issues of rel-
evance, in vivo and in vitro inconsistencies, and a lack of uniformity of operating
protocols. Because no single criterion is required for all probiotic uses, the best
way to determine a strain’s qualities is to conduct studies on a specific popula-
tion and physiologic function. Evidence supporting a strain’s probiotic efficacy
should thus be disseminated in medical journals or as peer-reviewed scientific
work, according to the Joint FAO/WHO Working Group. Negative results also add
to the overall body of evidence supporting probiotic efficacy and should likewise
be published.
Two important organs in the body, the stomach and the liver, are responsible for
lipoprotein production. The liver synthesizes bile which is then transported to the
gallbladder and utilized there prior to being transported to the intestine via a cystic
duct. When a fatty meal reaches the small intestine, bile salts assist in the emulsifica-
tion of the fats. This allows the fatty meal to be digested and absorbed in the intes-
tines. Next, in the epithelial cells of the gut, fatty acids, triglycerides, and cholesterol
mix and are covered by a layer of protein, called chylomicrons (Kaplan, Pesce, &
Kazmierczak, 2003). A system of lymphatic cells then absorbs the chylomicrons
after which they are released into the bloodstream. Chylomicrons are thus converted
to cholesterol and triglycerides upon reaching the liver. It is noteworthy that lipids
are immiscible with the bile salts in the intestines. Lipids descend to the ileum, where
absorption of a greater proportion of the bile salts occurs and then reabsorbed into
the bloodstream. The liver, therefore, is responsible for the recirculation of returned
bile salts that stay in the gallbladder, together with bile, to be employed in the afore-
mentioned process once again. The small intestine, however, does not absorb all of
the bile salts, and some of them move into the colon, where they are discarded along
with feces. By manufacturing bile salts from its cholesterol store, the liver compen-
sates for the loss of bile salts. In addition to dietary sources of cholesterol, liver cells
produce cholesterol and are thus another important source of the body’s cholesterol
pool. A variety of factors, including heredity and food, influence the liver’s ability to
generate cholesterol.
Dietary Modulation of Gut Microbiota 125
The enzyme BSH (choloylglycine hydrolase; EC 3.5.1.24) produces free BAs and
amino acid residues upon hydrolyzing glycine- and/or taurine-conjugated bile salts
(Figure 5.1), with a preference for glycol-conjugated bile salts over tauro-conjugated
bile salts. Bile salt conjugation significantly depends on the characteristics of the
host, and in mammals, the site of conjugation depends on the type of host. For exam-
ple, the microbial flora of Lactobacillus in the small intestine of mice are present at
the site where bile salt degeneration occurs, whereas, in humans, substantial flora
begin only at the end of the ileum and develop fully in the large intestine. Several
Dietary Modulation of Gut Microbiota 127
FIGURE 5.1 (A) The enzyme BSH hydrolyzes conjugated bile salts. (b) The involvement
of BSH in hypocholesterolemia and cholesterol as a mediator in the synthesis of new BAs. R:
Amino acid glycine or taurine. RDCA: Glyco- or tauro-deoxycholic acid, DCA: Deoxycholic
acid. Adapted from: Jones, Chen, Ouyang, Metz, and Prakash (2004) and Anandharaj,
Sivasankari, and Parveen Rani (2014).
researchers have proposed that the activity of BSH activity should be considered in
the probiotic selection criteria as they possess the ability to lower cholesterol. This
is because some probiotic microorganisms lacking this enzyme are not able to sig-
nificantly reduce cholesterol. Therefore, if bile secretion is an essential mechanism
for lowering cholesterol levels, the cultures used in in vivo tests should be selected
from the appropriate source. Numerous studies have demonstrated the distribution of
BSH activity in Bifidobacterium and Lactobacillus is genus, species, or even strain-
dependent. Most probiotic lactic acid bacteria originating from the human intestines
and feces have shown BSH activity. However, it is important to note that strains
with BSH activity isolated from the intestine or feces can also survive without this
enzyme and grow under bile salt conditions. BAs also stimulate cholesterol assimi-
lation through anaerobic development. The microbial metabolism of intestinal BAs
and the enterohepatic cycle defines the makeup of the pool of human BAs. Bile,
which is held in the gallbladder, is comprised primarily of conjugated BAs. Only 5%
of the BAs released in bile are reabsorbed in the terminal ileum and are recirculated
in the liver via the enterohepatic circulation to the large intestines, whereby 95%
of BAs are expelled in feces. While an array of colonic bacteria perform decon-
jugation activities, 7a-dehydroxylation appears to be limited to a small number of
intestinal bacteria. Consequently, the BAs profile expelled in feces and consisting
primarily of secondary BAs is highly dependent on gut microbial metabolism. The
pool measure, metabolite anatomy, and compartment fixations are important aspects
of the BA results because they relate to the normal gut microbiota composition and
128 The Gut Microbiome: Bench to Table
how it metabolizes BAs and influences host metabolic processes and obesity (Joyce
et al., 2014). The organisms use BAs to change the expression of genes controlled
by the farnesoid X receptor, resulting in a unique activation by the acids and their
metabolites (Sayin et al., 2013). Consequently, the more bile salts are secreted, the
more cholesterol will be removed from the bloodstream. Recently, the use of LAB
degenerated bile salts to reduce serum cholesterol levels in patients with hyperten-
sion and prevent hypertension in normal people has been receiving more attention.
The induction of such physiological effects was strongly connected to probiotic con-
sumption, prompting a further investigation to determine whether the effects were
due to native microbiota regulation or specific metabolic action of the consumed
probiotic strains (Hassan et al., 2019). Until recently, only one probiotic product
associated with claims of cardiovascular health has been approved in the market by
Health Canada. Cardioviva, which is also available in the United States and Europe,
contains two billion encapsulated Lactobacillus reuteri NCIMB 30242 and has been
clinically proven to lower LDL cholesterol levels by 11.6% in people with high cho-
lesterol levels (Jones, Martoni, & Prakash, 2012).
by the mice reduced total serum cholesterol levels by up to 25% and confirmed a
significant (P < 0.05) reduction in LDL, intermediate-density lipoprotein, and LDL
cholesterol levels. Probiotics must exhibit viability and growth in order to be able
to absorb cholesterol. In one study, L. plantarum 49 and L. plantarum 201 were
shown to lower TC serum levels after 14 days of bacterium consumption (da Costa
et al., 2019). In another study, L. mesenteroides subsp. mesenteroides KDK411 and
L. curvatus KFP419 were found to have potential hypercholesterolemia action in rats
through the absorption and excreting of cholesterol per evidence in the feces (Park,
Kim, Shin, Kim, & Whang, 2007).
Despite these results, some in vivo investigations have yielded contradictory
results. Since the cholesterol content of the feces of rats fed with L. acidophilus
ATCC 43121 (3 × 107 CFU/d) did not change from that of the control group, Park
et al. did not ascribe this mechanism to the hypocholesterolemic effect of the
probiotics. Moreover, adult individuals’ serum TC, LDL cholesterol, HDL choles-
terol, and TAG concentrations were not affected by treatment with L. rhamnosus
LC705 (two capsules/d containing 2×1010 CFU, for 4 weeks), even with a small
rise in serum cholesterol. As a result, we can conclude that the ability of bacteria
to assimilate cholesterol in the medium appears to be strain-specific and reliant
on the strain’s growth. The occurrence of the hypocholesterolemic effect can be
hindered if the strain can remove cholesterol but does not survive during passage
through the GI tract.
Binding of Cholesterol
Growing bacterial cells maintained a considerable quantity of cholesterol, and soni-
cation from cells of Bifidobacterium breve ATCC 15700 exuded more than 40%
of cholesterol. Even after several washes, detachment of the absorbed cholesterol
was impossible, indicating a strong binding potential between cholesterol and the
growing cells. Additionally, nongrowing Lactococcus cells have been confirmed to
have the capability of absorbing cholesterol in vitro; thus, the mechanism of choles-
terol removal has been studied from different perspectives. Probiotics endowed with
cholesterol-lowering properties possess the attribute of cholesterol-binding potential
in the small intestine. Nami et al (2019) reported that probiotic L. plantarum YS5
exhibited an elevated cholesterol-removal capacity by growing cells (84%) and mod-
erate cholesterol-removal capacity by resting (41.14%) cells and dead (32.71%) cells.
Moreover, L. fermentum lowered cholesterol levels in vitro, according to Pereira
and Gibson (2002). It has been suggested that cholesterol assimilation by gut cells
may lower the quantity of cholesterol available for intestinal absorption (Miremadi,
Ayyash, Sherkat, & Stojanovska, 2014; Pigeon, Cuesta, & Gilliland, 2002). Viable
as well as heat-killed cells were capable of cholesterol removal from growing media
by L. lactis subsp. lactis N7. Viable cells were also able to substantially eliminate
more cholesterol than dead cells (Kimoto-Nira et al., 2007). L. gasseri strains have
been confirmed to eliminate cholesterol in vitro by adhering to the cellular surface,
although this capacity seemed to be growth- and strain-specific. The capacity of pro-
biotics to eliminate cholesterol throughout various growth conditions, notwithstand-
ing, bolstered the aforementioned result (Kimoto, Ohmomo, & Okamoto, 2002).
Dietary Modulation of Gut Microbiota 131
Other Mechanisms
Metabolic products such as SCFAs synthesized by probiotics have a pivotal role in
cholesterol removal. SCFAs serve as ligands to activate peroxisome proliferator-
activated receptors (PPARs). Angiopoietin-like protein 4 (ANGPTL4), a lipoprotein
lipase (LPL) inhibitor, is produced by brown and white fat in the liver and intestines
and is regulated by PPARγ (Hoda, 2011). According to Sheril et al. (2013), buty-
rates activate PPARs predominantly, followed by propionate, and finally acetate. As a
result of PPAR activation and the upregulation of ANGPTL4 levels, SCFAs decrease
LPL activities. This action suppresses fat storage by regulating fatty acid oxidation
in muscle and adipocytes. Therefore, to have a hypocholesterolemic impact, the pro-
biotic microorganisms must create enough propionate to counteract the effects of
acetate on cholesterogenesis. For example, L. plantarum MA2 (1011 cells/d) incor-
porated into a cholesterol-enriched diet and fed to Sprague–Dawley rats for 5 weeks
exhibited a higher fecal propionate concentration than that of the control group.
Cholesterol can be converted to coprostanol (5-cholestan-3-ol) and coprostanone
in small concentrations. These metabolites have a poor intestinal absorption rate
and are excreted in the feces with a decrease in cholesterol absorption. The cho-
lesterol reductase enzyme must be present in the cholesterol-lowering probiotics in
order to convert cholesterol. This enzyme is present in probiotic bacteria such as
Lactobacillus bulgaricus FTCC 0411, L. acidophilus FTCC 0291, L. acidophilus
ATCC 314, L. casei 1311 FTDC ATCC 393, B. bifidum PRL2010, and Eubacterium
coprostanoligenes ATCC 51222 (Gérard, 2014; H-S Lye et al., 2010; Zanotti et al.,
2015). There is limited information in the literature regarding the strains that are
positive for this enzyme; however, the activity of this enzyme is strain-specific.
The Niemann-Pick C1-Like 1 (NPC1L1) protein is found in the small intestines
on the brush surface membrane of enterocytes and plays an integral role in cho-
lesterol absorption (Jia, Betters, & Yu, 2011). In a study by Huang et al., hyper-
cholesterolemic rats were treated with L. acidophilus ATCC 4356 for 28 days, and
it was found that NPC1L1 levels in segments of the jejunum and the duodenum
were considerably decreased (P < 0.05) (Huang et al., 2013). This was corroborated
and confirmed by Duval et al. that activators of the liver X receptor (LXR) sup-
pressed the expression of the NPC1L1 gene in the gut. Interestingly, in mammals,
cholesterol metabolism is primarily regulated by LXRs. LXRα- and LXRβ- are the
two kinds of LXR that have been identified so far and regulate the metabolism of
lipids and carbohydrates. These LXRs are nuclear receptors and constitute mem-
bers of a superfamily that functions as ligand-activated transcription factors. Huang
and Zheng (2010) found that the expression of LXR-α and LXR-β was upregulated
by the L. acidophilus ATCC 4356 strain, whereas the expression of NPC1L1 was
downregulated in a dose- and time-dependent manner in Caco-2. In addition, L
plantarum LRCC 5273 effectively decreased hypercholesterolemia in mice by acti-
vating hepatic and intestinal LXR-, leading to BA excretion and increased fecal
cholesterol in the small intestine (Heo et al., 2018).
Choline, phosphatidylcholine, L-carnitine, and betaine were metabolized by the
intestinal microbiota to trimethylamine (TMA), which is then oxidized to trimethyl-
amine N-oxide (TMAO) by hepatic flavin monooxygenases (FMO3) (Zhou et al., 2020).
132 The Gut Microbiome: Bench to Table
TMAO levels in the blood have been linked to negative outcomes in CVD patients such
as chronic heart failure and coronary heart disease. Consequently, attention to TMAO
levels could be a potentially useful predictive indicator for unfavorable cardiac events
in patients with chronic heart failure following myocardial infarction. In addition, the
formation of gut microbiota-dependent TMAO is inhibited and has proven to be a via-
ble method for atherosclerosis therapy (Z. Wang et al., 2015). However, it is yet unclear
which gut microorganisms play the most important role in CVD, and the precise pro-
cesses involved need to be investigated further (Molinero et al., 2019).
With regard to intestinal disorders, it is essential to exclude, from the diet, foods
that impair the motor function of the intestine. In other words, the food must be
mechanically, chemically, and thermally sparing. It is thus necessary to reduce fat
consumption as the quantity and quality of fats impact their degree of digestion by
the body. Therapeutic diets are also required to provide the necessary amount of
energy and protein. In addition, to address intestinal disorders, therapeutic diets
should include a substantial amount of vitamins and minerals (Cardona, Andrés-
Lacueva, Tulipani, Tinahones, & Queipo-Ortuño, 2013).
The primary bacterial groups that make up the intestinal microflora of the
human body are Firmicutes, Bacteroidetes, Proteobacteria, Verrucomicrobia, and
Actinobacteria. The amount and species composition of the intestinal microflora of
a healthy person varies with age and the composition of the food, and most of these
bacteria are commensal (they coexist with the human body without causing disease).
Children are born with a sterile digestive tract, which begins to colonize with micro-
organisms after a few hours. Enterobacteria and Bifidobacteria are the first settlers.
However, a healthy stomach and the proximal part of the small intestine contain very
few organisms, which is due to the bactericidal action of hydrochloric acid in gas-
tric juice. The colon, on the other hand, is dominated by anaerobic microorganisms,
while the small intestine is predominantly aerobic and anaerobic. Bacteria in the
intestinal lumen break down different types of sterols and steroids. The bacteria also
convert the cholic acid of bile salts into deoxycholate, break down toxins, produce
biotin and pantothenic acid, stimulate the immune system, and cause the fermenta-
tion of cellulose (Schmidt, Raes, & Bork, 2018).
Many factors affect the degree of microbial presence in the gut. Factors that
influence the ecosystem of the intestinal microbiota include the mode of birth, the
mother’s microbiota, breastfeeding, exposure to environmental bacteria, intake
of antibiotics/probiotics, and diet (Figure 5.2). It was found that in terms of com-
position, variety, and functional competencies, the microbiota of 2- to 3-year-old
children is close to that of adults (Yang, Ye, Yan, He, & Xing, 2019). It is believed
that the composition of the microbiota after three years of age is complete and
functional. Moreover, eating habits and overall healthy lifestyles could generally
promote a healthy intestinal microbiota (Yang et al., 2019).
There is a strong link between nutrient intake and the composition of intestinal
microbiota. It is known that intestinal microorganisms can affect the synthesis of
specific vitamins or the breakdown of certain nutrients, that is, the effects of micro-
biota on nutrition. Moreover, the intestinal microbiota also differs among people
from different latitudes and is also buttressed by environmental and genetic factors,
as well as the use and administration of antibiotics (Goodrich et al., 2016). Nutrition,
however, is suggested as the main reason for the differences in the composition of the
gut microbiota between people.
Studies have shown that different food components are used by different bacterial
communities and ensure the colonization of this community; in this way, the domi-
nant bacterial species can be formed depending on the diet. Among the nutrients,
carbohydrates, fiber, protein, fat, phytochemicals, and vitamins play a significant
role in creating the intestinal microbiota (Duncan et al., 2007). Studies examining
the effects of a high-fat diet on the intestinal fat microbiota have shown that such a
diet reduces microbial diversity and increases the number of Bacteroides, Alistipe,
and Bilophila. High-fat diets increase the concentration of SCFAs in the feces com-
pared to low-fat diets and significantly reduce the levels of bifidobacteria (Heiman
& Greenway, 2016). Phytochemicals are biologically active substances derived from
plants that give plants their color, odor, and natural stability. It is known that a diet
rich in fruits, vegetables, grains, and legumes reduces the risk of various diseases.
The prophylactic effects, therefore, of these foods are partly due to antioxidants in
their composition. The beneficial health effects of foods with probiotics such as dairy
products have been well studied. For example, in recent years, we have seen increas-
ing evidence that probiotics may play an essential role in the proper functioning of
gut health and digestion, urogenital tract, immune system, respiratory features, and
allergies, and can significantly combat various infectious diseases (Yang et al., 2019).
LI, symptoms of constipation, stimulating the immune system, allergies, cancer, pre-
vention of various types of diarrhea, and regulation of cholesterol levels (Liu, Tran,
& Rhoads, 2018). There are many studies on probiotic use in diarrhea due to the
use of antibiotics, and inflammatory bowel diseases (IBDs) such as ulcerative colitis
(UC), chronic illness, spastic irritable bowel syndrome, and necrotizing enterocolitis
(Ryma, Samer, Soufli, Rafa, & Touil-Boukoffa, 2021). Oligosaccharides cannot be
hydrolyzed or absorbed in the small intestine. However, they can be fermented by
Lactobacillus spp. and Bifidobacterium spp. and have prebiotic properties. In the
food industry, the most widely used oligosaccharides are the following: fructooligo-
saccharides, galactooligosaccharides (GOS), transgalactooligosaccharides, xylooli-
gosaccharides, gentio, lactulose, lactosaccharose, inulin, isomaltooligosaccharides,
preharisaccharides, and salt. Inulin has also been widely used in food in recent years
and can be fermented by Lactobacillus spp. and Bifidobacterium spp., and has prebi-
otic characteristics (Liu et al., 2018).
Cardiovascular Disease
Globally, CVD is extensive, and the death toll attributed to this disease is increas-
ing. Estimates by the WHO confirm that approximately 23 million people world-
wide will have CVD by 2023, affecting up to 22% of the global economy. As
human societies progress, lifestyle changes, and mobility declines, anomalies such
as impaired insulin production or insulin resistance have led to increased levels
of triglycerides, total cholesterol, low-density lipoprotein (LDL-C), and decreased
LPL-C lipoprotein. High (HDL-C) cholesterol rises in the blood, thus posing an
elevated CVD health risk in susceptible individuals. In addition, cholesterol oxi-
dized by the formation of arterial plaques is a major cause of coronary heart disease.
These factors, in combination with widespread smoking, contribute to the bulk of
critical modifiable risk factors, which are responsible for approximately half of all
deaths globally. Aortic and other peripheral vascular blood vessel inflammation is
also a less prevalent health concern linked to atherosclerosis. According to Lee et
al. (2012), CVD is attributed as one of the major precursors of death and sickness in
the world, accounting for 29% of overall worldwide mortality (Lee et al., 2013). In
addition to these variable basic factors, ACD mortality is influenced by extremely
widespread physiologic and metabolic changes. Dyslipidemias, obesity, high blood
sugar, hypertension, and insulin resistance are the most common diseases (Ramos,
Esteves, Prates, Moreno, & Santos, 2022). For example, people with hypercholes-
terolemia are three times more susceptible to having a heart attack than those with
a normal lipid profile.
Liver Disease
During the first years of use of lactulose in hepatic encephalopathy, the colon pH
decreases as a result of the bacterial metabolism of lactulose, which facilitates the
proliferation of Lactobacillus and other acidophilic bacteria that can tolerate acid
while preventing the formation of ammonia. Lactobacillus is thought to avoid the
proliferation of the responsible acidophobic, proteolytic bacteria. Some researchers
contend that bacterial proliferation in the colon increases with lactulose, that the
increased bacterial population uses ammonia as a source of nitrogen or that there
136 The Gut Microbiome: Bench to Table
Dental Diseases
The activity of probiotics on the health of the oral cavity and teeth is related to
the ability of probiotics to produce an antimicrobial agent against oral pathogens.
Probiotic bacteria can release various antimicrobial substances such as organic acid,
hydrogen peroxide, carbon dioxide, diacetyl, bacteriocin, and adhesion inhibitors,
which have high antimicrobial potential. Probiotics can also adhere to the surface of
the teeth as well as the mouth surface. This is an essential point with regard to the
long-term probiotic effects of bacteria. Lactobacilli can adhere to hydroxyapatite
on teeth. Most adhesion experiments for probiotic purposes were based on the daily
consumption of cheese and yogurt. Another vital ability of probiotics is their effect
on the immune system. Probiotics are thought to have mechanisms that can alter
the host’s immune system. Dendritic cells scattered on the mucosa surface play a
crucial role in the pre-identification of bacteria and activation of T-cell responses.
Depending on the signal, either immune tolerance or an active immune response
against a particular antigen is established in the dendritic cells. Due to the ability of
probiotic bacteria to produce various antimicrobial substances and adhesion inhibi-
tors that affect the oral flora, probiotics can significantly affect the environmental
conditions necessary for the survival of bacteria by altering the pH of the environ-
ment and the redox potential (Haukioja, 2010).
Dietary Modulation of Gut Microbiota 137
Gastrointestinal Diseases
Inflammatory bowel Disease
Inflammatory bowel disease is an inflammatory, intermittent, chronic GI tract dis-
ease that recurs spontaneously. IBD is a term used to describe a group of complex
disorders of the GI tract. IBDs include two main forms: Crohn’s disease (CD) and UC,
which share similar clinical symptoms. Symptoms and signs of the disease include
stool changes in CD; however, recurrence is observed when stool change disappears.
Inflammation of the mucosa is reduced when broad-spectrum enteric-coated antibi-
otics are used for UC. Triggering environmental conditions, genetic predisposition,
and intestinal bacteria plays a role in the occurrence of IBD. Studies in humans and
animals have shown that the percentage of beneficial microorganisms such as probi-
otics decreases the incidence of IBD (Lakatos, 2006).
Manipulation of the intestinal flora has become an interesting approach to pre-
venting and treating IBD. Different results were obtained with other probiotic strains
and their combined use in experimental models of colitis (Ryma et al., 2021).
Ulcerative colitis: Another well-known chronic IBD is UC whereby inflamma-
tion occurs as a result of abnormal reactions of the immune system. Many studies
have shown that probiotics effectively maintain remission in UC. When fecal suspen-
sions of healthy individuals are given to patients with UC by enema, the symptoms
can be relieved and remission achieved.
In placebo-controlled studies, the incidence of relapse was found to be signifi-
cantly reduced in probiotic users. With the establishment of remission, NF-κB,
IL-1β, TNF-α decreased, and IL-10 increased (Baumgart & Carding, 2007; Ryma
et al., 2021).
Crohn’s disease: This is another IBD that leads to inflammation of the digestive
tract. The distribution of bacteria in the gut of individuals with CD is very different
from that of normal individuals. For this reason, although it was thought possible
to approximate the flora of normal individuals through the use of probiotics, it was
not possible to make a definite assessment of the results obtained (Nagao-Kitamoto,
Kitamoto, Kuffa, & Kamada, 2016). CD is characterized by a discontinuous pattern,
affecting the entire GI tract. In CD patients, the inflammation is transmural with
large ulcerations and granulomas.
Several clinical trials and experimental studies displayed the role of Saccharomyces
boulardii as a good biotherapeutic agent for preventing and treating several GI dis-
eases including CD. S. boulardii stimulates effects that resemble the protective
effects of the normal healthy gut flora (Kelesidis & Pothoulakis, 2012).
Pouchitis is inflammation occurring in the ileal region after ileal-anal anasto-
mosis, in which the irregularities in the intestinal flora act as a triggering factor.
In a study investigating the efficacy of a probiotic preparation (VSL#3) containing
5 × 10(11) per gram of viable lyophilized bacteria of four strains of Lactobacilli,
three strains of Bifidobacteria, and one strain of Streptococcus salivarius subsp.
thermophilus, it was confirmed that this probiotic preparation was as efficient as
compared to a placebo in the maintenance of remission of chronic pouchitis. The
results showed that fecal concentration of Lactobacilli, Bifidobacteria, and S. ther-
mophilus increased significantly from baseline levels only in the VSL#3-treated
138 The Gut Microbiome: Bench to Table
group. Thus, oral administration of the new probiotic preparation was effective in
preventing flare-ups of chronic pouchitis (Gionchetti et al., 2000). While 85% of
study subjects had a decrease in disease effects after 9 months, there was evidence of
disease recurrence in all individuals in the placebo group.
Cancer Treatment
Diet plays a role in the development of colorectal cancer. A diet rich in meat and
animal fats, and low in fiber, changes the distribution of colon flora. As Clostridium
and Bacteroides strains increase, Bifidobacterium strains decrease. Because flora
and the immune system play a role in tumor development, it is thought that, theoreti-
cally, prebiotics and probiotics that regulate flora can also be used to prevent tumor
development. For example, lactulose causes a decrease in 7-α dehydroxylase activity
through changes in the bacterial flora of the colon. 7-α dehydroxylase is the enzyme
that causes the formation of secondary BAs. Secondary BAs are considered cocar-
cinogens. In addition, secondary BAs enter the enterohepatic circulation and form
a continuous cycle, affecting BAs and cholesterol synthesis. The production of 7-α
dehydroxylase and other potentially toxic enzymes of bacterial origin is reduced after
administration of lactulose. Thus, lactulose administration helps in the reduction
of toxic substances such as ammonia, which is also a known potential carcinogen.
One hypothesis is that butyrate formed in the colon using lactulose has a DNA-
protective impact. In addition to the many positive effects on health, probiotics also
have anticancer, tumor-suppressant, and therapeutic effects (Pitsillides et al., 2021).
Studies investigating probiotic–cancer interactions have demonstrated the capabili-
ties of probiotics, such as the cancer-preventing effects. Probiotics enhance the host’s
immune response, deteriorate the structure of potentially carcinogenic compounds,
create qualitative and quantitative changes in the intestinal flora, and have antimu-
tagenic effects on the colon. Among the positive effects of probiotic action could be
the ability of probiotics to produce anticancer compounds, alteration of metabolic
Dietary Modulation of Gut Microbiota 139
Lactose Intolerance
Lactose intolerance is one of the most common forms of food intolerance and occurs
when lactase activity is reduced in the small intestine lining. LI is characterized by
GI symptoms, including vomiting, diarrhea, flatulence, and abdominal pain, which
are caused by the fermentation of unabsorbed lactose in the colon. The severity of
LI and the symptoms mentioned above can vary considerably from individual to
individual. Lactase deficiency can be primary, secondary, or congenital. The most
common form is primary lactase deficiency, a consequence of lactase deficiency
characterized by a progressive decrease in lactase activity (Leis, de Castro, de
Lamas, Picáns, & Couce, 2020).
If there is no or low lactase activity in the gut, undegraded lactose disturbs the
osmotic balance and causes the accumulation of fluids and electrolytes in the gut. In
enlarged intestines, motility increases, and diarrhea occurs.
Lactose, which reaches the colon without being broken down, is fermented by
bacteria, resulting in hydrogen, methane, and carbon dioxide. Yogurt is the most
superior probiotic product due to its beneficial effects on LI (Gyawali et al., 2020;
Ibrahim et al., 2021). Because some microorganisms in yogurt contain the enzyme
lactase, they break down lactose before it reaches the colon and prevents LI symp-
toms and signs.
People who have digestive problems with lactose can tolerate yogurt because there
is beta-galactosidase activity in the bacteria found in yogurt. S. thermophilus and
Lactobacillus delbrueckii subsp. bulgaricus contain the enzyme beta-galactosidase
(lactase), which improves the digestion of lactose. Bacteria in yogurt metabolize lac-
tose by releasing bacterial lactase due to lysis through the effect of bile salts in the
small intestine after digestion. In addition, because fermented dairy products are
darker than milk, their passage time through the GI tract is longer which allows for
better digestion.
140 The Gut Microbiome: Bench to Table
Yogurt and other fermented products of dairy origin are generally considered by
consumers to be good sources of health-promoting organisms (Tremblay & Panahi,
2017). Yogurt by definition is a fermented cultured milk product containing two
strains of live bacteria, namely Lactobacillus delbrueckii subsp. bulgaricus and
Streptococcus thermophilus. These microbes should be at least 107cfu/g and live at
the time of consumption (Savaiano & Hutkins, 2021). Often other genera of probiotic
Lactobacillus and Bifidobacterium strains are also added into yogurt for additional
health benefits. For instance, yogurt-based diets have been linked to a decreased risk
of bladder cancer, reduced weight gain, and a reduced risk of metabolic syndrome
Dietary Modulation of Gut Microbiota 141
TABLE 5.2
Intervention Studies and Impact of Cultured Food Products on Gut Microbiota
Foods Study Population Intervention Findings References
Yogurt Thirty-eight Four-week ingestion of Reduced E. coli: Yang and
children with H. probiotics-containing Bifidobacterium spp. ratio in Sheu
pylori infection yogurt children infected in H. pylori (2012)
Yogurt Twenty-three Six months of daily Bifidobacterium and total C. Wang
drink healthy ingestion of a probiotic Lactobacillus increased, and et al.
preschool (Lactobacillus casei strain Enterobacteriaceae, (2015)
children Shirota) containing drink Staphylococcus, and
Clostridium perfringens
decreased
Yogurt Fifty-eight Four-week commercial Viable counts of fecal Savard
volunteers: yogurt consumption lactobacilli were et al.
healthy women supplemented with significantly higher, and (2011)
and men aged Bifidobacterium animalis those of enterococci were
between 18 and subsp. lactis and significantly lower
55 years Lactobacillus acidophilus
Kefir Forty-five (23 400 mL/day kefir was Lactobacillus bacterial load of Yılmaz,
male and 22 administered twice a day feces of all subjects in the Dolar, and
female) patients to the patients for 4 weeks, treatment group was Özpınar
completed the which contains between 104 and 109 CFU/g, (2019)
study Lactobacillus bacteria and the first and last
(treatment group, 25 measurements were
patients) statistically significant
Kefir Eighty-two Patients were given 250 mL Results showed a 14-day Bekar et al.
patients with of kefir twice daily (triple regimen of triple therapy (2011)
symptoms of therapy + kefir, n = 46) or with kefir is more effective
dyspepsia and H. 250 mL of milk in achieving H.
pylori infection containing placebo (triple pylori eradication than triple
therapy + placebo, n = 36) therapy alone
for 14 days
Probiotic Six healthy female The volunteers consumed Bacteroidetes increased Unno et al.
milk volunteers, two servings of drink during ingestion and (2015)
20–24 years of daily for 3 weeks, and decreased again after the end
age then stopped consuming of ingestion. Firmicutes
for an additional 3 weeks changed in the opposite way
of the study period
Probiotic Thirty-two Thirty-two subjects Probiotic milk product Veiga et al.
milk women (aged consumed (125 g/serving) containing dairy starters (2014)
20–69 years) twice a day either the and Bifidobacterium
probiotic milk (n = 17) or animalis potentiates colonic
an acidified milk product SCFA production and
(n = 15) for 4 weeks decreases abundance of a
pathobiont Bilophila
wadsworthia compared to a
milk product
(Continued)
142 The Gut Microbiome: Bench to Table
(Tremblay & Panahi, 2017). Over a century ago, Elie Metchnikoff proposed that
by modifying the intestinal microbiome with bacteria present in yogurt, one’s
health could be improved (Mackowiak, 2013). Odamaki et al. (2012) assessed 420
healthy people and found that frequent yogurt consumption containing probiotic
Bifidobacterium longum BB536 reduced the amount of enterotoxigenic Bacteroides
fragilis in the gut. These results also corroborated the conclusion that endotoxin-
producing bacteria are significantly suppressed by the habitual consumption of
yogurt (Odamaki et al., 2012). A four-week intervention study with yogurt contain-
ing probiotic bacteria reduced the intestinal Escherichia coli: Bifidobacterium spp.
ratio in children infected with Helicobacter pylori when compared with the control.
This study showed reduced H. pylori loads and elevated serum IgA concentrations
in infected children (Yang & Sheu, 2012). Donovan and Shamir (2014) confirmed
the enormous health benefits associated with yogurt consumption including the
prevention of GI diseases, as well as the maintenance of healthy gut microbiota
(Donovan & Shamir, 2014). For example, consumption of probiotic yogurt drink
fermented with the strain, L. casei Shirota, showed an increase in the total fecal
Lactobacillus count and decreased Enterobacteriaceae and Staphylococcus counts
Dietary Modulation of Gut Microbiota 143
(Tsuji et al., 2014; C. Wang et al., 2015). The consumption of fermented yogurt
containing Bifidobacterium lactis and Lactobacillus fermentum strains was also
shown to elevate fecal Bifidobacterium and Lactobacillus counts (Ahmed, Prasad,
Gill, Stevenson, & Gopal, 2007; Ibrahim et al., 2021; Savard et al., 2011; West et al.,
2011). It has been reported that consumption of yogurt is associated with an increase
in Lactobacillus gasseri and a decrease in Enterobacteriaceae and Staphylococcus
levels. Another important health benefit associated with yogurt is the alleviation
of LI symptoms due to the expression and release of β-galactosidase by the yogurt
cultures (Gyawali et al., 2020; Ibrahim et al., 2021).
Kefir is a cultured dairy product produced by the action of yeasts, lactic acid bac-
teria, and acetic acid bacteria fermentation of lactose in milk (Guzel-Seydim, Kok-
Tas, Greene, & Seydim, 2011). Recently, kefir drink has raised increased interest
among health-conscious consumers due to its superior health benefits. The impact
of kefir on gut microbiota was investigated by Santos et al. (2003), where isolated
kefir strains exhibited adhesion properties to the human enterocyte-like Cco-2 cells
indicating a potential ability to colonize the human gut (Santos, San Mauro, Sanchez,
Torres, & Marquina, 2003). Some studies on kefir strains have shown an increase
in Lactobacillus, Bifidobacterium, and Lactococcus populations, and a decrease in
Proteobacteria and Enterobacteriaceae counts in the gut microbiota (Kim et al.,
2015). In another human study (n = 45), Lactobacillus kefiri was identified in fecal
samples after 800 mL/day of kefir was consumed for 4 weeks. The total Lactobacillus
count in stool samples was significantly higher than as compared to the control sam-
ples in patients with CD (Bekar, Yilmaz, & Gulten, 2011). Likewise, a double-blind
randomized controlled trial investigated the impact of consumption of 500 mL/day
kefir on the eradication of Helicobacter pylori in patients with dyspepsia. The results
showed that the control group (250 mL/day milk) had a lower eradication rate of H.
pylori compared to the kefir group which had significantly higher levels of H. pylori
(Bekar et al., 2011). Kefir also helped to improve LI symptoms, improve the immune
system, lower cholesterol, and was shown to have anticarcinogenic and antimuta-
genic properties (Guzel-Seydim et al., 2011).
an increased number of Lactobacillus spp. (Inoguchi et al., 2012) in ten healthy adults.
Similarly, consumption of extracts of fermented plants for 8 weeks by 22 hypercho-
lesterolemic patients was found to increase bifidobacteria and Lactobacillus spp. and
decrease E. coli and C. perfringens counts (Chiu et al., 2017).
CONCLUSION
In the last few decades, probiotic consumption has gained much attention due to its
association with health benefits. Additionally, there has been increasingly widespread
knowledge regarding the role of nutrition and diet in supporting a healthy gut micro-
biota. In vitro, experimental, and clinical researches have all demonstrated the syn-
ergistic effect of diet and probiotic use on enhancing the modulatory mechanism of
the gut microbiota in specifically addressing human health and disease challenges.
Consequently, it has also been established from the literature that cultured and fer-
mented functional food products exert a positive impact on the overall metabolic
functionality of the human system. The mechanism of probiotic action against dis-
ease-causing organisms helps to enhance the performance of the gut microbiota by the
production of metabolites that support proper coordination and hormonal imbalance.
Diet and nutrition are thus important key factors that influence the composition of the
gut microbiota and serve as a driving force in promoting human health and wellness.
As presented in this chapter, the importance of diet and cultured food products in the
modulation of the gut microbiome cannot be overemphasized. Thus, the blending of
diet, nutrition, and probiotic consumption as a part of an individual’s lifestyle will
result in a natural and therapeutic approach to disease prevention via the resultant
modulatory effect on the gut microbiota without compromising the immune system.
REFERENCES
Ahmed, M., Prasad, J., Gill, H., Stevenson, L., & Gopal, P. (2007). Impact of consumption of
different levels of bifidobacterium lactis HN019 on the intestinal microflora of elderly
human subjects. Journal of Nutrition Health and Aging, 11(1), 26.
Aller, R., De Luis, D., Izaola, O., Conde, R., Gonzalez Sagrado, M., Primo, D., . . . Gonzalez, J.
(2011). Effect of a probiotic on liver aminotransferases in nonalcoholic fatty liver dis-
ease patients: a double blind randomized clinical trial. European Review for Medical
Pharmacological Sciences, 15(9), 1090–1095.
Altonsy, M. O., Andrews, S. C., & Tuohy, K. M. (2010). Differential induction of apoptosis
in human colonic carcinoma cells (Caco-2) by atopobium, and commensal, probiotic
and enteropathogenic bacteria: mediation by the mitochondrial pathway. International
Journal of Food Microbiology, 137(2–3), 190–203.
An, S. J., Kim, J. Y., Kim, I. S., Adhikari, B., Yu, D. Y., Kim, J. A., . . . Cho, K. K. (2019).
Modulation of intestinal microbiota by supplementation of fermented kimchi in rats.
Journal of Life Science, 29(9), 986–995.
Anandharaj, M., Sivasankari, B., & Parveen Rani, R. (2014). Effects of probiotics, prebiot-
ics, and synbiotics on hypercholesterolemia: a review. Chinese Journal of Biology,
2014, 1-7.
Antoun, M., Hattab, Y., Akhrass, F.-A., & Hamilton, L. D. (2020). Uncommon pathogen,
lactobacillus, causing infective endocarditis: case report and review. Case Reports in
Infectious Diseases, 2020, 1–4.
Anusha Siddiqui, S., Redha, A. A., Esmaeili, Y., & Mehdizadeh, M. (2022). Novel insights on
extraction and encapsulation techniques of elderberry bioactive compounds. Critical
Reviews in Food Science and Nutrition, 1–16.
Ayivi, R. D., Gyawali, R., Krastanov, A., Aljaloud, S. O., Worku, M., Tahergorabi, R.,...
& Ibrahim, S. A. (2020). Lactic acid bacteria: food safety and human health applica-
tions. Dairy, 1(3), 202–232.
148 The Gut Microbiome: Bench to Table
Baumgart, D. C., & Carding, S. R. (2007). Inflammatory bowel disease: cause and immuno-
biology. The Lancet, 369(9573), 1627–1640.
Behrouzi, A., Mazaheri, H., Falsafi, S., Tavassol, Z. H., Moshiri, A., & Siadat, S. D. (2020).
Intestinal effect of the probiotic Escherichia coli strain Nissle 1917 and its OMV.
Journal of Diabetes & Metabolic Disorders, 19(1), 597–604.
Bekar, O., Yilmaz, Y., & Gulten, M. (2011). Kefir improves the efficacy and tolerability of triple
therapy in eradicating Helicobacter pylori. Journal of Medicinal Food, 14(4), 344–347.
Bhat, B., & Bajaj, B. K. (2020). Multifarious cholesterol lowering potential of lactic acid
bacteria equipped with desired probiotic functional attributes. 3 Biotech, 10(5), 1–16.
Bordoni, A., Amaretti, A., Leonardi, A., Boschetti, E., Danesi, F., Matteuzzi, D., . . . Rossi, M.
(2013). Cholesterol-lowering probiotics: in vitro selection and in vivo testing of bifido-
bacteria. Applied Microbiology and Biotechnology, 97(18), 8273–8281.
Brashears, M., Gilliland, S., & Buck, L. (1998). Bile salt deconjugation and cholesterol removal
from media by Lactobacillus casei. Journal of Dairy Science, 81(8), 2103–2110.
Byakika, S., Mukisa, I. M., Byaruhanga, Y. B., & Muyanja, C. (2019). A review of criteria
and methods for evaluating the probiotic potential of microorganisms. Food Reviews
International, 35(5), 427–466.
Cardona, F., Andrés-Lacueva, C., Tulipani, S., Tinahones, F. J., & Queipo-Ortuño, M. I.
(2013). Benefits of polyphenols on gut microbiota and implications in human health.
The Journal of Nutritional Biochemistry, 24(8), 1415–1422.
Cheng, I.-C., Shang, H.-F., Lin, T.-F., Wang, T.-H., Lin, H.-S., & Lin, S.-H. (2005). Effect
of fermented soy milk on the intestinal bacterial ecosystem. World Journal of
Gastroenterology: WJG, 11(8), 1225.
Chiu, H.-F., Chen, Y.-J., Lu, Y.-Y., Han, Y.-C., Shen, Y.-C., Venkatakrishnan, K., & Wang, C.-K.
(2017). Regulatory efficacy of fermented plant extract on the intestinal microflora and
lipid profile in mildly hypercholesterolemic individuals. Journal of Food and Drug
Analysis, 25(4), 819–827.
Choi, H.-J., Lee, N.-K., & Paik, H.-D. (2015). Health benefits of lactic acid bacteria iso-
lated from kimchi, with respect to immunomodulatory effects. Food Science and
Biotechnology, 24(3), 783–789.
Coton, M., Pawtowski, A., Taminiau, B., Burgaud, G., Deniel, F., Coulloumme-Labarthe, L., . . .
Coton, E. (2017). Unraveling microbial ecology of industrial-scale Kombucha fermen-
tations by metabarcoding and culture-based methods. FEMS Microbiology Ecology,
93(5), 1–16.
Croft, J., Cresanta, J., Webber, L., Srinivasan, S., Freedman, D., Burke, G., & Berenson, G.
(1988). Cardiovascular risk in parents of children with extreme lipoprotein cholesterol
levels: the Bogalusa Heart Study. Southern Medical Journal, 81(3), 341–349, 353.
da Costa, W. K. A., Brandão, L. R., Martino, M. E., Garcia, E. F., Alves, A. F., de Souza, E. L., . . .
Vidal, H. (2019). Qualification of tropical fruit-derived Lactobacillus plantarum strains
as potential probiotics acting on blood glucose and total cholesterol levels in Wistar
rats. Food Research International, 124, 109–117.
Das, G., Paramithiotis, S., Sivamaruthi, B. S., Wijaya, C. H., Suharta, S., Sanlier, N., . . .
Patra, J. K. (2020). Traditional fermented foods with anti-aging effect: a concentric
review. Food Research International, 134, 109269.
Dashkevicz, M. P., & Feighner, S. D. (1989). Development of a differential medium for bile
salt hydrolase-active Lactobacillus spp. Applied and Environmental Microbiology,
55(1), 11–16.
De Preter, V., Vanhoutte, T., Huys, G., Swings, J., De Vuyst, L., Rutgeerts, P., & Verbeke, K.
(2007). Effects of Lactobacillus casei Shirota, Bifidobacterium breve, and oligo-
fructose-enriched inulin on colonic nitrogen-protein metabolism in healthy humans.
American Journal of Physiology-Gastrointestinal and Liver Physiology, 292(1),
G358–G368.
Dietary Modulation of Gut Microbiota 149
Derrien, M., & van Hylckama Vlieg, J. E. (2015). Fate, activity, and impact of ingested bacte-
ria within the human gut microbiota. Trends in microbiology, 23(6), 354–366.
Donovan, S. M., & Shamir, R. (2014). Introduction to the yogurt in nutrition initiative and the
First Global Summit on the health effects of yogurt. The American Journal of Clinical
Nutrition, 99(5), 1209S–1211S.
Dufresne, C., & Farnworth, E. (2000). Tea, Kombucha, and health: a review. Food Research
International, 33(6), 409–421.
Duncan, S. H., Belenguer, A., Holtrop, G., Johnstone, A. M., Flint, H. J., & Lobley, G. E.
(2007). Reduced dietary intake of carbohydrates by obese subjects results in decreased
concentrations of butyrate and butyrate-producing bacteria in feces. Applied and
Environmental Microbiology, 73(4), 1073–1078.
Ebringer, L., Ferenčík, M., & Krajčovič, J. (2008). Beneficial health effects of milk and fer-
mented dairy products. Folia Microbiologica, 53(5), 378–394.
Esmaeili, Y., Paidari, S., Baghbaderani, S. A., Nateghi, L., Al-Hassan, A., & Ariffin, F.
(2021). Essential oils as natural antimicrobial agents in postharvest treatments of fruits
and vegetables: a review. Journal of Food Measurement and Characterization, 16,
507–522.
El-Saadony, M. T., Alagawany, M., Patra, A. K., Kar, I., Tiwari, R., Dawood, M. A., Dhama, K.,
Abdel-Latif, H. M. (2021). The functionality of probiotics in aquaculture: an overview.
Fish & Shellfish Immunology, 1(117), 36–52.
Esmaeili, Y., Zamindar, N., Paidari, S., Ibrahim, S. A., & Mohammadi Nafchi, A. (2021).
The synergistic effects of aloe vera gel and modified atmosphere packaging on the
quality of strawberry fruit. Journal of Food Processing and Preservation, 45(12),
e16003.
FAO & WHO. (2002). Joint FAO/WHO working group report on drafting guidelines for the
evaluation of probiotics in food. Geneva: World Health Organization.
Fuller, R. (1989). Probiotics in man and animals. Journal of Applied Bacteriology, 66,
365–378.
Fuller, R. (1999). Probiotics for farm animals. Probiotics: A Critical Review, 15–22.
Galie, N., Manes, A., Negro, L., Palazzini, M., Bacchi-Reggiani, M. L., & Branzi, A. (2009).
A meta-analysis of randomized controlled trials in pulmonary arterial hypertension.
European Heart Journal, 30(4), 394–403.
Ganguly, N., Bhattacharya, S., Sesikeran, B., Nair, G., Ramakrishna, B., Sachdev, H., . . .
Kathuria, S. (2011). ICMR-DBT guidelines for evaluation of probiotics in food. The
Indian Journal of Medical Research, 134(1), 22.
Gentile, C. L., & Weir, T. L. (2018). The gut microbiota at the intersection of diet and human
health. Science, 362(6416), 776–780.
Gérard, P. (2014). Metabolism of cholesterol and bile acids by the gut microbiota. Pathogens,
3(1), 14–24.
Gionchetti, P., Rizzello, F., Venturi, A., Brigidi, P., Matteuzzi, D., Bazzocchi, G., . . .
Campieri, M. (2000). Oral bacteriotherapy as maintenance treatment in patients with
chronic pouchitis: a double-blind, placebo-controlled trial. Gastroenterology, 119(2),
305–309.
Goodrich, J. K., Davenport, E. R., Beaumont, M., Jackson, M. A., Knight, R., Ober, C., . . .
Ley, R. E. (2016). Genetic determinants of the gut microbiome in UK twins. Cell Host
& Microbe, 19(5), 731–743.
Guzel-Seydim, Z. B., Kok-Tas, T., Greene, A. K., & Seydim, A. C. (2011). Functional proper-
ties of kefir. Critical Reviews in Food Science and Nutrition, 51(3), 261–268.
Gyawali, R., Oyeniran, A., Zimmerman, T., Aljaloud, S. O., Krastanov, A., & Ibrahim, S. A.
(2020). A comparative study of extraction techniques for maximum recovery of
β-galactosidase from the yogurt bacterium Lactobacillus delbrueckii ssp. bulgaricus.
Journal of Dairy Research, 87(1), 123–126.
150 The Gut Microbiome: Bench to Table
Han, K., Bose, S., Wang, J., Kim, B. S., Kim, M. J., Kim, E. J., & Kim, H. (2015). Contrasting
effects of fresh and fermented kimchi consumption on gut microbiota composition and
gene expression related to metabolic syndrome in obese Korean women. Molecular
Nutrition & Food Research, 59(5), 1004–1008.
Hassan, A., Din, A. U., Zhu, Y., Zhang, K., Li, T., Wang, Y., . . . Wang, G. (2019). Updates in
understanding the hypocholesterolemia effect of probiotics on atherosclerosis. Applied
Microbiology and Biotechnology, 103(15), 5993–6006.
Haukioja, A. (2010). Probiotics and oral health. European Journal of Dentistry, 4(3), 348–355.
Heiman, M. L., & Greenway, F. L. (2016). A healthy gastrointestinal microbiome is dependent
on dietary diversity. Molecular Metabolism, 5(5), 317–320.
Heo, W., Lee, E. S., Cho, H. T., Kim, J. H., Lee, J. H., Yoon, S. M., . . . Kim, Y.-J. (2018).
Lactobacillus plantarum LRCC 5273 isolated from Kimchi ameliorates diet-induced
hypercholesterolemia in C57BL/6 mice. Bioscience, Biotechnology, and Biochemistry,
82(11), 1964–1972.
Hoda, M. (2011). Probiotics bacteria from Egyptian infants cause cholesterol removal in
media and survive in yoghurt. Food and Nutrition Sciences, 2, 150–155.
Holscher, H. D. (2017). Dietary fiber and prebiotics and the gastrointestinal microbiota. Gut
microbes, 8(2), 172–184.
Huang, Y., & Zheng, Y. (2010). The probiotic Lactobacillus acidophilus reduces choles-
terol absorption through the down-regulation of Niemann-Pick C1-like 1 in Caco-2
cells. British Journal of Nutrition, 103(4), 473–478.
Huang, Y., Wang, X., Wang, J., Wu, F., Sui, Y., Yang, L., & Wang, Z. (2013). Lactobacillus
plantarum strains as potential probiotic cultures with cholesterol-lowering activity.
Journal of Dairy Science, 96(5), 2746–2753.
Ibrahim, S. A., Gyawali, R., Awaisheh, S. S., Ayivi, R. D., Silva, R. C., Subedi, K., . . .
Krastanov, A. (2021). Fermented foods and probiotics: an approach to lactose intoler-
ance. Journal of Dairy Research, 88(3), 357–365.
Inoguchi, S., Ohashi, Y., Narai-Kanayama, A., Aso, K., Nakagaki, T., & Fujisawa, T. (2012).
Effects of non-fermented and fermented soybean milk intake on faecal microbiota and
faecal metabolites in humans. International Journal of Food Sciences and Nutrition,
63(4), 402–410.
Jia, L., Betters, J. L., & Yu, L. (2011). Niemann-pick C1-like 1 (NPC1L1) protein in intestinal
and hepatic cholesterol transport. Annual Review of Physiology, 73, 239–259.
Jones, M., Martoni, C., & Prakash, S. (2012). Cholesterol lowering and inhibition of ste-
rol absorption by Lactobacillus reuteri NCIMB 30242: a randomized controlled trial.
European Journal of Clinical Nutrition, 66(11), 1234–1241.
Jones, M. L., Chen, H., Ouyang, W., Metz, T., & Prakash, S. (2004). Microencapsulated genet-
ically engineered Lactobacillus plantarum 80 (pCBH1) for bile acid deconjugation and
its implication in lowering cholesterol. Journal of Biomedicine and Biotechnology,
2004(1), 61–69.
Joseph, N., Vasodavan, K., Saipudin, N. A., Yusof, B. N. M., Kumar, S., & Nordin, S. A.
(2019). Gut microbiota and short-chain fatty acids (SCFAs) profiles of normal and
overweight school children in Selangor after probiotics administration. Journal of
Functional Foods, 57, 103–111.
Joyce, S. A., MacSharry, J., Casey, P. G., Kinsella, M., Murphy, E. F., Shanahan, F., . . . Gahan,
C. G. (2014). Regulation of host weight gain and lipid metabolism by bacterial bile acid
modification in the gut. Proceedings of the National Academy of Sciences, 111(20),
7421–7426.
Jung, Y., Kim, I., Mannaa, M., Kim, J., Wang, S., Park, I., . . . Seo, Y.-S. (2019). Effect of
Kombucha on gut-microbiota in mouse having non-alcoholic fatty liver disease. Food
science and biotechnology, 28(1), 261–267.
Dietary Modulation of Gut Microbiota 151
Kaplan, L. A., Pesce, A. J., & Kazmierczak, S. C. (2003). Clinical chemistry: theory, analy-
sis, correlation (Vol. 1): Mosby Incorporated, St. Louis, MO.
Kasprzak-Drozd, K., Oniszczuk, T., Stasiak, M., & Oniszczuk, A. (2021). Beneficial effects of
phenolic compounds on gut microbiota and metabolic syndrome. International Journal
of Molecular Sciences, 22(7), 3715.
Kato-Kataoka, A., Nishida, K., Takada, M., Kawai, M., Kikuchi-Hayakawa, H., Suda, K., . . .
Matsuki, T. (2016). Fermented milk containing Lactobacillus casei strain Shirota pre-
serves the diversity of the gut microbiota and relieves abdominal dysfunction in healthy
medical students exposed to academic stress. Applied and Environmental Microbiology,
82(12), 3649–3658.
Kechagia, M., Basoulis, D., Konstantopoulou, S., Dimitriadi, D., Gyftopoulou, K.,
Skarmoutsou, N., & Fakiri, E. M. (2013). Health benefits of probiotics: a review.
ISRN Nutrition, 2013, 481651. https://doi.org/10.5402/2013/481651
Kelesidis, T., & Pothoulakis, C. (2012). Efficacy and safety of the probiotic Saccharomyces
boulardii for the prevention and therapy of gastrointestinal disorders. Therapeutic
Advances in Gastroenterology, 5(2), 111–125.
Khare, A., & Gaur, S. (2020). Cholesterol-lowering effects of Lactobacillus species. Current
Microbiology, 77(4), 638–644.
Kil, J.-H., Jung, K.-O., Lee, H.-S., Hwang, I.-K., Kim, Y.-J., & Park, K.-Y. (2004). Effects
of kimchi on stomach and colon health of Helicobacter pylori-infected volunteers.
Preventive Nutrition and Food Science, 9(2), 161–166.
Kim, D.-H., Chon, J.-W., Kim, H.-S., Yim, J.-H., Kim, H., & Seo, K.-H. (2015). Rapid detec-
tion of Lactobacillus kefiranofaciens in kefir grain and kefir milk using newly devel-
oped real-time PCR. Journal of Food Protection, 78(4), 855–858.
Kimoto-Nira, H., Mizumachi, K., Nomura, M., Kobayashi, M., Fujita, Y., Okamoto, T., . . .
Ohmomo, S. (2007). Lactococcus sp. as potential probiotic lactic acid bacteria. Japan
Agricultural Research Quarterly: JARQ, 41(3), 181–189.
Kimoto, H., Ohmomo, S., & Okamoto, T. (2002). Cholesterol removal from media by lacto-
cocci. Journal of Dairy Science, 85(12), 3182–3188.
Kumar, M., Nagpal, R., Kumar, R., Hemalatha, R., Verma, V., Kumar, A., . . . Jain, S. (2012).
Cholesterol-lowering probiotics as potential biotherapeutics for metabolic diseases.
Experimental Diabetes Research, 2012, 1-14.
Lakatos, P. L. (2006). Recent trends in the epidemiology of inflammatory bowel diseases: up
or down? World Journal of Gastroenterology: WJG, 12(38), 6102.
Le, B., & Yang, S.-H. (2019). Effect of potential probiotic Leuconostoc mesenteroides FB111
in prevention of cholesterol absorption by modulating NPC1L1/PPARα/SREBP-2 path-
ways in epithelial Caco-2 cells. International Microbiology, 22(2), 279–287.
Lee, H., Lee, S., Hwang, I., Park, Y., Yoon, S., Han, K., . . . Yim, H. (2013). Prevalence,
awareness, treatment and control of hypertension in adults with diagnosed diabetes:
the Fourth Korea National Health and Nutrition Examination Survey (KNHANES IV).
Journal of Human Hypertension, 27(6), 381–387.
Leis, R., de Castro, M.-J., de Lamas, C., Picáns, R., & Couce, M. L. (2020). Effects of pre-
biotic and probiotic supplementation on lactase deficiency and lactose intolerance: a
systematic review of controlled trials. Nutrients, 12(5), 1487.
Lim, H., Oh, S., Yu, S., & Kim, M. (2021). Isolation and characterization of probiotic Bacillus
subtilis MKHJ 1-1 possessing L-asparaginase activity. Applied Sciences, 11(10), 4466.
Liong, M., & Shah, N. (2005). Acid and bile tolerance and cholesterol removal ability of lac-
tobacilli strains. Journal of Dairy Science, 88(1), 55–66.
Liu, M., Xie, W., Wan, X., & Deng, T. (2020). Clostridium butyricum modulates gut
microbiota and reduces colitis associated colon cancer in mice. International
Immunopharmacology, 88, 106862.
152 The Gut Microbiome: Bench to Table
Liu, Y., Tran, D. Q., & Rhoads, J. M. (2018). Probiotics in disease prevention and treatment.
The Journal of Clinical Pharmacology, 58, S164–S179.
Lye, H.-S., Rahmat-Ali, G. R., & Liong, M.-T. (2010). Mechanisms of cholesterol removal by
lactobacilli under conditions that mimic the human gastrointestinal tract. International
Dairy Journal, 20(3), 169–175.
Lye, H.-S., Rusul, G., & Liong, M.-T. (2010). Removal of cholesterol by lactobacilli via incor-
poration and conversion to coprostanol. Journal of Dairy Science, 93(4), 1383–1392.
Ma, E. L., Choi, Y. J., Choi, J., Pothoulakis, C., Rhee, S. H., & Im, E. (2010). The antican-
cer effect of probiotic Bacillus polyfermenticus on human colon cancer cells is medi-
ated through ErbB2 and ErbB3 inhibition. International Journal of Cancer, 127(4),
780–790.
Mackowiak, P. A. (2013). Recycling Metchnikoff: probiotics, the intestinal microbiome and
the quest for long life. Frontiers in Public Health, 1, 52.
Marco, M. L., Heeney, D., Binda, S., Cifelli, C. J., Cotter, P. D., Foligné, B., . . . Pihlanto, A.
(2017). Health benefits of fermented foods: microbiota and beyond. Current Opinion in
Biotechnology, 44, 94–102.
McAuliffe, O., Cano, R. J., & Klaenhammer, T. R. (2005). Genetic analysis of two bile salt
hydrolase activities in Lactobacillus acidophilus NCFM. Applied and Environmental
Microbiology, 71(8), 4925–4929.
Meroni, M., Longo, M., & Dongiovanni, P. (2019). The role of probiotics in nonalcoholic fatty
liver disease: a new insight into therapeutic strategies. Nutrients, 11(11), 2642.
Miremadi, F., Ayyash, M., Sherkat, F., & Stojanovska, L. (2014). Cholesterol reduction mech-
anisms and fatty acid composition of cellular membranes of probiotic Lactobacilli and
Bifidobacteria. Journal of Functional Foods, 9, 295–305.
Miyazaki, M., Yoshitomi, H., Miyakawa, S., Uesaka, K., Unno, M., Endo, I., . . . Shimada,
K. (2015). Clinical practice guidelines for the management of biliary tract cancers
2015: the 2nd English edition. Journal of Hepato‐Biliary‐Pancreatic Sciences, 22(4),
249–273.
Mohammed, O. A., Simon, C. A, Kieran, M. T. (2010). Differential induction of apoptosis
in human colonic carcinoma cells (Caco-2) by Atopobium, and commensal, probiotic
and enteropathogenic bacteria: Mediation by the mitochondrial pathway. International
Journal of Food Microbiology, 137(2–3), 190–203.
Molinero, N., Ruiz, L., Sánchez, B., Margolles, A., & Delgado, S. (2019). Intestinal bacte-
ria interplay with bile and cholesterol metabolism: implications on host physiology.
Frontiers in Physiology, 10, 185.
Nagao-Kitamoto, H., Kitamoto, S., Kuffa, P., & Kamada, N. (2016). Pathogenic role of the gut
microbiota in gastrointestinal diseases. Intestinal research, 14(2), 127.
Nair, D. V., & Kollanoor Johny, A. (2018). Characterizing the antimicrobial function of
a dairy-originated probiotic, Propionibacterium freudenreichii, against multidrug-
resistant Salmonella enterica serovar Heidelberg in turkey poults. Frontiers in
Microbiology, 9, 1475.
Nami, Y., Haghshenas, B., Bakhshayesh, R. V., Jalaly, H. M., Lotfi, H., Eslami, S., & Hejazi,
M. A. (2018). Novel autochthonous lactobacilli with probiotic aptitudes as a main
starter culture for probiotic fermented milk. LWT, 98, 85–93. https://doi.org/10.1016/j.
lwt.2018.08.035
Nami, Y., Vaseghi Bakhshayesh, R., Mohammadzadeh Jalaly, H., Lotfi, H., Eslami, S., &
Hejazi, M. A. (2019). Probiotic properties of Enterococcus isolated from artisanal dairy
products. Frontiers in Microbiology, 10, 300.
Odamaki, T., Sugahara, H., Yonezawa, S., Yaeshima, T., Iwatsuki, K., Tanabe, S., . . . Xiao,
J.-z. (2012). Effect of the oral intake of yogurt containing Bifidobacterium longum
BB536 on the cell numbers of enterotoxigenic Bacteroides fragilis in microbiota.
Anaerobe, 18(1), 14–18.
Dietary Modulation of Gut Microbiota 153
Pandey, K. B., & Rizvi, S. I. (2009). Plant polyphenols as dietary antioxidants in human
health and disease. Oxidative medicine and cellular longevity, 2(5), 270–278.
Park, Y.-H., Kim, J.-G., Shin, Y.-W., Kim, S.-H., & Whang, K.-Y. (2007). Effect of dietary
inclusion of Lactobacillus acidophilus ATCC 43121 on cholesterol metabolism in rats.
Journal of Microbiology and Biotechnology, 17(4), 655–662.
Pereira, D. I., & Gibson, G. R. (2002). Cholesterol assimilation by lactic acid bacteria and bifido-
bacteria isolated from the human gut. Applied and Environmental Microbiology, 68(9),
4689–4693.
Pigeon, R., Cuesta, E., & Gilliland, S. (2002). Binding of free bile acids by cells of yogurt
starter culture bacteria. Journal of Dairy Science, 85(11), 2705–2710.
Pitsillides, L., Pellino, G., Tekkis, P., & Kontovounisios, C. (2021). The effect of perioperative
administration of probiotics on colorectal cancer surgery outcomes. Nutrients, 13(5), 1451.
Ramos, C. L., Esteves, E. A., Prates, R. P., Moreno, L. G., & Santos, C. S. (2022). Probiotics
in the prevention and management of cardiovascular diseases with focus on dyslipid-
emia. In Dwivedi, M. K., Amaresan, N., Sankaranarayanan, A., Kemp, H. E., (Eds.),
Probiotics in the prevention and management of human diseases (pp. 337–351):
Elsevier, Academic Press, USA.
Rodríguez-Nogales, A., Algieri, F., Garrido-Mesa, J., Vezza, T., Utrilla, M. P., Chueca, N., . .
. Gálvez, J. (2018). Intestinal anti-inflammatory effect of the probiotic Saccharomyces
boulardii in DSS-induced colitis in mice: impact on microRNAs expression and gut
microbiota composition. The Journal of Nutritional Biochemistry, 61, 129–139.
Rong, S., Hu, X., Zhao, S., Zhao, Y., Xiao, X., Bao, W., & Liu, L. (2017). Procyanidins
extracted from the litchi pericarp ameliorate atherosclerosis in ApoE knockout mice:
their effects on nitric oxide bioavailability and oxidative stress. Food & Function, 8(11),
4210–4216.
Ryma, T., Samer, A., Soufli, I., Rafa, H., & Touil-Boukoffa, C. (2021). Role of probiotics and
their metabolites in inflammatory bowel diseases (IBDs). Gastroenterology Insights,
12(1), 56–66.
Sadeghi-Aliabadi, H., Mohammadi, F., Fazeli, H., & Mirlohi, M. (2014). Effects of
Lactobacillus plantarum A7 with probiotic potential on colon cancer and normal cells
proliferation in comparison with a commercial strain. Iranian Journal of Basic Medical
Sciences, 17(10), 815.
Santos, A., San Mauro, M., Sanchez, A., Torres, J., & Marquina, D. (2003). The antimicrobial
properties of different strains of Lactobacillus spp. isolated from kefir. Systematic and
applied Microbiology, 26(3), 434–437.
Savaiano, D. A., & Hutkins, R. W. (2021). Yogurt, cultured fermented milk, and health: a
systematic review. Nutrition Reviews, 79(5), 599–614.
Savard, P., Lamarche, B., Paradis, M.-E., Thiboutot, H., Laurin, É., & Roy, D. (2011). Impact
of Bifidobacterium animalis subsp. lactis BB-12 and, Lactobacillus acidophilus LA-5-
containing yoghurt, on fecal bacterial counts of healthy adults. International Journal
of Food Microbiology, 149(1), 50–57.
Sayin, S. I., Wahlström, A., Felin, J., Jäntti, S., Marschall, H.-U., Bamberg, K., . . . Bäckhed, F.
(2013). Gut microbiota regulates bile acid metabolism by reducing the levels of tauro-beta-
muricholic acid, a naturally occurring FXR antagonist. Cell Metabolism, 17(2), 225–235.
Schmidt, T. S., Raes, J., & Bork, P. (2018). The human gut microbiome: from association to
modulation. Cell, 172(6), 1198–1215.
Shabbir, U., Rubab, M., Daliri, E. B.-M., Chelliah, R., Javed, A., & Oh, D.-H. (2021).
Curcumin, quercetin, catechins and metabolic diseases: the role of gut microbiota.
Nutrients, 13(1), 206.
Shastri, M. D., Chong, W. C., Vemuri, R., Martoni, C. J., Adhikari, S., Bhullar, H., . . . Eri, R. D.
(2020). Streptococcus thermophilus UASt-09 upregulates goblet cell activity in colonic
epithelial cells to a greater degree than other probiotic strains. Microorganisms, 8(11), 1758.
154 The Gut Microbiome: Bench to Table
Shehata, M. G., El-Sahn, M. A., El Sohaimy, S. A., & Youssef, M. M. (2019). In vitro assess-
ment of hypocholesterolemic activity of Lactococcus lactis subsp. lactis. Bulletin of the
National Research Centre, 43(1), 1–10.
Sheril, A., Lange, K., Amolo, T., Grinstead, J. S., Haakonsson, A. K., Szalowska, E., ... &
Kersten, S. (2013). Short-chain fatty acids stimulate angiopoietin-like 4 synthesis in
human colon adenocarcinoma cells by activating peroxisome proliferator-activated
receptor γ. Molecular and Cellular Biology, 33(7), 1303–1316.
Shimizu, M., Hashiguchi, M., Shiga, T., Tamura, H.-o., & Mochizuki, M. (2015). Meta-
analysis: effects of probiotic supplementation on lipid profiles in normal to mildly
hypercholesterolemic individuals. PloS One, 10(10), e0139795.
Sidira, M., Galanis, A., Ypsilantis, P., Karapetsas, A., Progaki, Z., Simopoulos, C., &
Kourkoutas, Y. (2010). Effect of probiotic-fermented milk administration on gastroin-
testinal survival of Lactobacillus casei ATCC 393 and modulation of intestinal micro-
bial flora. Microbial Physiology, 19(4), 224–230.
Suganya, K., Son, T., Kim, K.-W., & Koo, B.-S. (2021). Impact of gut microbiota: how it could
play roles beyond the digestive system on development of cardiovascular and renal dis-
eases. Microbial Pathogenesis, 152, 104583.
Tamang, J. P., Shin, D.-H., Jung, S.-J., & Chae, S.-W. (2016). Functional properties of micro-
organisms in fermented foods. Frontiers in Microbiology, 7, 578.
Tao, J.-h., Duan, J.-a., Jiang, S., Qian, Y.-y., & Qian, D.-w. (2016). Biotransformation and
metabolic profile of buddleoside with human intestinal microflora by ultrahigh-perfor-
mance liquid chromatography coupled to hybrid linear ion trap/orbitrap mass spectrom-
eter. Journal of Chromatography B, 1025, 7–15.
Tarrah, A., dos Santos Cruz, B. C., Sousa Dias, R., da Silva Duarte, V., Pakroo, S., Licursi
de Oliveira, L., . . . Oliveira de Paula, S. (2021). Lactobacillus paracasei DTA81, a
cholesterol‐lowering strain having immunomodulatory activity, reveals gut microbi-
ota regulation capability in BALB/c mice receiving high‐fat diet. Journal of Applied
Microbiology, 131(4), 1942–1957.
Thumu, S. C. R., & Halami, P. M. (2020). In vivo safety assessment of Lactobacillus fer-
mentum strains, evaluation of their cholesterol‐lowering ability and intestinal microbial
modulation. Journal of the Science of Food and Agriculture, 100(2), 705–713.
Tremblay, A., & Panahi, S. (2017). Yogurt consumption as a signature of a healthy diet and
lifestyle. The Journal of Nutrition, 147(7), 1476S–1480S.
Tsuji, H., Chonan, O., Suyama, Y., Kado, Y., Nomoto, K., Nanno, M., & Ishikawa, F. (2014).
Maintenance of healthy intestinal microbiota in women who regularly consume probi-
otics. International Journal of Probiotics & Prebiotics, 9(1/2), 31.
Unno, T., Choi, J.-H., Hur, H.-G., Sadowsky, M. J., Ahn, Y.-T., Huh, C.-S., . . . Cha, C.-J.
(2015). Changes in human gut microbiota influenced by probiotic fermented milk inges-
tion. Journal of Dairy Science, 98(6), 3568–3576.
Valdes, A. M., Walter, J., Segal, E., & Spector, T. D. (2018). Role of the gut microbiota in
nutrition and health. BMJ, 361, 36–44
Vasiljevic, T., & Shah, N. P. (2008). Probiotics—from Metchnikoff to bioactives. International
Dairy Journal, 18(7), 714–728.
Veiga, P., Pons, N., Agrawal, A., Oozeer, R., Guyonnet, D., Brazeilles, R., . . . Whorwell, P. J.
(2014). Changes of the human gut microbiome induced by a fermented milk product.
Scientific Reports, 4(1), 1–9.
Wang, C., Nagata, S., Asahara, T., Yuki, N., Matsuda, K., Tsuji, H., . . . Yamashiro, Y.
(2015). Intestinal microbiota profiles of healthy pre-school and school-age children
and effects of probiotic supplementation. Annals of Nutrition and Metabolism, 67(4),
257–266.
Dietary Modulation of Gut Microbiota 155
Wang, L., Guo, M.-J., Gao, Q., Yang, J.-F., Yang, L., Pang, X.-L., & Jiang, X.-J. (2018). The
effects of probiotics on total cholesterol: a meta-analysis of randomized controlled tri-
als. Medicine, 97(5).
Wang, Z., Roberts, A. B., Buffa, J. A., Levison, B. S., Zhu, W., Org, E., . . . Culley, M. K.
(2015). Non-lethal inhibition of gut microbial trimethylamine production for the treat-
ment of atherosclerosis. Cell, 163(7), 1585–1595.
West, N. P., Pyne, D. B., Cripps, A. W., Hopkins, W. G., Eskesen, D. C., Jairath, A., . . .
Fricker, P. A. (2011). Lactobacillus fermentum (PCC®) supplementation and gastroin-
testinal and respiratory-tract illness symptoms: a randomised control trial in athletes.
Nutrition Journal, 10(1), 1–11.
Xu, R.-y., Wan, Y.-p., Fang, Q.-y., Lu, W., & Cai, W. (2011). Supplementation with probiot-
ics modifies gut flora and attenuates liver fat accumulation in rat nonalcoholic fatty
liver disease model. Journal of Clinical Biochemistry and Nutrition, 1108190104–
1108190104, 1–6.
Yadav, B. S., Yadav, A. K., Singh, S., Singh, N. K., & Mani, A. (2019). Methods in metage-
nomics and environmental biotechnology. In Gothandam, K. M., Ranjan, S., Dasgupta, N.,
Lichtfouse, E. (Eds.), Nanoscience and biotechnology for environmental applications
(pp. 85–113): Springer.
Yang, Q., Liang, Q., Balakrishnan, B., Belobrajdic, D. P., Feng, Q. J., & Zhang, W. (2020). Role
of dietary nutrients in the modulation of gut microbiota: a narrative review. Nutrients,
12(2), 381.
Yang, B., Ye, C., Yan, B., He, X., & Xing, K. (2019). Assessing the influence of dietary history
on gut microbiota. Current Microbiology, 76(2), 237–247.
Yang, Y. J., & Sheu, B. S. (2012). Probiotics‐containing yogurts suppress Helicobacter pylori
load and modify immune response and intestinal microbiota in the Helicobacter pylori‐
infected children. Helicobacter, 17(4), 297–304.
Yılmaz, İ., Dolar, M. E., & Özpınar, H. (2019). Effect of administering kefir on the changes
in fecal microbiota and symptoms of inflammatory bowel disease: a randomized con-
trolled trial. The Turkish Journal of Gastroenterology, 30(3), 242.
Zanotti, I., Turroni, F., Piemontese, A., Mancabelli, L., Milani, C., Viappiani, A., . . . Elviri, L.
(2015). Evidence for cholesterol-lowering activity by Bifidobacterium bifidum PRL2010
through gut microbiota modulation. Applied Microbiology and Biotechnology, 99(16),
6813–6829.
Zhou, X., Jin, M., Liu, L., Yu, Z., Lu, X., & Zhang, H. (2020). Trimethylamine N‐oxide and
cardiovascular outcomes in patients with chronic heart failure after myocardial infarc-
tion. ESC Heart Failure, 7(1), 189–194.
6 Dietary Modulation of
the Gut Microbiota by
Prebiotics and Other
Dietary Nutrients and
Relevant Health Effect(s)
Alison Lacombe and Vivian C.H. Wu
United States Department of Agriculture
CONTENTS
Introduction............................................................................................................. 157
Types of Prebiotics.................................................................................................. 158
Carbohydrate Prebiotics..................................................................................... 159
Oligosaccharides........................................................................................... 159
Human Milk Oligosaccharides...................................................................... 159
Polysaccharides............................................................................................. 161
Inulin............................................................................................................. 162
Non-Carbohydrate Prebiotics.................................................................................. 163
Polyphenols........................................................................................................ 163
Short-Chain Fatty Acids..................................................................................... 164
Metabolism of Nutrients by the Gut Microbiota..................................................... 164
Effect of Prebiotic on Immunity........................................................................ 164
Prebiotics as Functional Foods............................................................................... 166
Novel Prebiotic Formulations............................................................................ 168
Food-Processing Effects on Probiotics and Prebiotics....................................... 169
Factors That Influence Probiotic Viability......................................................... 170
Conclusion.............................................................................................................. 173
References............................................................................................................... 173
INTRODUCTION
A significant advancement in gastrointestinal microbiology realized the impact diet
extends on health status and how gut constituent affects the host’s health (David
et al. 2014). Within the last 30 years, science has transitioned from viewing the gas-
trointestinal tract (GIT) as an inert tube to a flourishing ecosystem akin to vibrant
and diverse dynamic communities in the Amazon rainforest. Similar to the deepest
recesses of planet Earth, little is known about the gut community and, more impor-
tantly, what inputs keep it thriving. This chapter focuses on the relationship between
the gut microbiota, the host diet, and the resulting symbiotic relationship. Evidence
accumulated by in vitro work and recent evidence from in vivo studies and human
clinical trials collectively indicate that phytochemicals, fiber, and other nutrients
demonstrate prebiotic capabilities (David et al. 2014). Parsing through the panoply of
research requires a deep look into the chemical structure of prebiotics in the gut and
the metabolic orchestration of the host gut microbiome.
Prebiotic compounds cannot be digested in the human stomach and small intestine
and instead pass through to the large intestine. Prebiotics enrich a particular category
of bacteria in the large intestine that is proven beneficial to gut health. While numer-
ous bacteria provide health benefits to their host, the families of Bifidobacteriaceae
and Lactobacillaceae specifically dominate the published research even though they
merely comprise less than 0.01% of the intestinal microbiome. Before the 1960s,
Bifidobacteria was known as Lactobacillus bifidus because it was observed to ferment
carbohydrates into lactic acid. It was later discovered that Bifidobacteria employs
a unique fructose-6-phosphate phosphoketolase pathway to ferment carbohydrates.
This characteristic feature has allowed Bifidobacteria to dominate the scientific con-
versation around prebiotics. Advances in whole genome sequencing technology (see
the previous chapters) challenge the conventional consensus of probiotic definition.
However, this progress is impeded because many of these species cannot be cultured
and subsequently enumerated and characterized outside their host.
TYPES OF PREBIOTICS
As far back as 1921, several experiments described the enrichment of Lactobacilli
in humans following the consumption of carbohydrates (Gibson et al. 2017). The
first formal definition for prebiotics was established in 1995 as a “nondigestible food
ingredient that beneficially affects the host by selectively stimulating the growth and
activity of one or a limited number of bacteria already resident in the colon” (Cheplin
and Rettger 2011). The definition of prebiotics has expanded over the years as
advances in metagenomics and metabolomics have made it possible to take a clearer
snapshot of the gastrointestinal environment (Gibson et al. 2017). Genomic technolo-
gies have expanded the definitions of nondigestible foods, beneficial bacteria, and
the site of interaction (Gibson et al. 2017). This definition continues to evolve, and
emerging evidence demonstrates that synergistic activities between microorganisms
are not limited to the GIT and nondigestible carbohydrates (Gibson et al. 2017). Here,
we will define prebiotics as a food source established to sustain microorganisms. The
result is a symbiotic relationship with sustained benefits to the host. Here, we discuss
the specific nutrients and microorganisms that support such a beneficial relationship.
The popularity of gut health in the wellness industry has allowed the rapid expan-
sion of products that support beneficial microbes. Several products on the market of
undefined composition claim prebiotic capabilities (Hume, Nicolucci, and Reimer
2017). Here, we approach the nondigestible food ingredients/metabolites with an
evidence-based approach demonstrating stimulation of the growth and metabolism
of beneficial gut microbes. Prebiotics must confer specific positive changes in gut
Dietary Modulation of the Gut Microbiota by Prebiotics 159
The most common prebiotics with a strong basis of evidence are indigestible carbo-
hydrates (i.e., dietary fiber, such as inulin and oligosaccharides) (Collins and Reid
2016). Although prebiotics are derived from digestible fiber, not all fiber types are
prebiotics (Collins and Reid 2016). Bacteria differ in their carbohydrate processing
capabilities, and most gut microbiota members selectively metabolize mono- and
oligosaccharides. Complex polysaccharides often require multiple microorganisms
with specialized enzymatic repertoires for complete digestion to utilizable energy
units. Most commercially available prebiotic carbohydrates come from plants; how-
ever, emerging research on animals, algae, and bacteria provides promising alterna-
tives (Gurpilhares et al. 2019).
Numerous food compounds are resistant to digestion and are used for fermentation
by gut microflora. Saccharolytic fermentation occurs with nondigestible carbohy-
drates and favors the growth of some specific bacteria genera such as Bifidobacterium
and Lactobacillus. These saccharolytic bacteria produce short-chain fatty acids
(SCFA) in the colon, namely acetate, propionate, or butyrate, which contribute calo-
ries, absorbed by the gut, and extend the putative effect on systemic inflammation.
Oligosaccharides
Oligosaccharides are relatively small chains ranging from three to ten simple
saccharides, eliciting a variety of functions in cell biology and host immunity.
Oligosaccharides are classified by their monosaccharide constituents and linkages to
lipids (glycolipids) or amino acids (glycoproteins). Not all oligosaccharides occur as
components of glycoproteins or glycolipids. Raffinose is a storage or transport car-
bohydrate in plants, and maltodextrins result from the microbial breakdown of larger
polysaccharides such as starch or cellulose (Fernandes et al. 2017).
In this section, a particular focus is placed on specific examples of oligosaccha-
rides particularly interested in those with a prebiotic function, namely fructooligo-
saccharides (FOS) and galactooligosaccharides (GOS). FOS elicit between 30% and
50% of the sweetness that sucrose confers. They are different from fructans and
inulin, which can be precursors of oligosaccharides, and demonstrate a much higher
degree of polymerization than that of FOS and GOS (Figure 6.1).
FIGURE 6.2 General structure of milk oligosaccharides and a comparison between human
and bovine oligosaccharides.
The advancement of metabolomics and proteomics has allowed for the in-depth
analysis of HMO constituents. The findings from these studies suggest that each
mother’s HMO composition constitutes a unique fingerprint rather than being some-
thing homogeneous across the population (Bode 2012; Azad et al. 2018). There
are several maternal determinants of the final composition of human breast milk.
Lifestyle, exercise, and health status are predictors of the relative abundance of
fats, proteins, and oligosaccharides overall (Moossavi et al. 2019; Azad et al. 2018;
Addison et al. 2019). However, the relative abundance of individual oligosaccharides
is driven primarily by genetics. Principle coordinate analysis of 10,000 human milk
samples taken from six continents shows two distinct clusters (Azad et al. 2018).
The differences have been linked to a single nucleotide polymorphism (SNP) that
encodes for an enzyme called fucosyltransferase II (fucII) that adds L-fucose to the
oligosaccharide chain forming a specific α-1,2 linkage (Bode 2012). These SNPs
map out geographically, as shown by genome-wide sequence analysis and transcrip-
tomics. For example, 90% of Peruvian women have an active fucII, but that number
goes down to 65% on the African continent (Kober, Zehe, and Bode 2013; Bode
2015). Several additional SNPs are associated with the formation of different HMO
patterns, and more information can be found at the University of California, San
Diego, MOMI Core Institute.
Polysaccharides
Polysaccharides are heterogeneous macromolecules composed of monosaccharides
linked by glycosidic linkages. Typical examples of biologically essential polysac-
charides are starches (glucose polymers), a mixture of amylose and amylopectin,
162 The Gut Microbiome: Bench to Table
and the animal counterpart glycogen, cellulose, chitin, and pectin (O’Connor et al.
2017). As with the previous section, most of the research on prebiotic polysaccha-
rides focuses on their bifidogenic effects. Research is still probing the “tip of the
iceberg” regarding polysaccharides and gut microbiota. Not all polysaccharides
enter the large intestine intact, but the ones that do tend to have bifidogenic effects
(O’Connor et al. 2017). Our current understanding suggests that this is because of the
“resistant” nature of the molecule itself and the enzymatic repertoire of bacteria like
Bifidobacteria. This assumption is not corroborated in human models when investi-
gating in depth the types of bacteria affected by dietary changes.
Inulin
Inulin is a generic term that covers all linear fructans with beta (2−1) fructosyl-fructose
glycosidic bonds (Kelly 2008). The beta configuration of the bonds between fructose
monomers and inulin-type fructans resists enzymatic hydrolysis by human salivary
constituents and small intestinal digestive enzymes allowing for them to pass to the
large intestine intact. As a result, inulin-type fructans are indigestible and fermented
in the colon (Kelly 2008). Inulin is present in over 3,000 vegetables and extensively
present in plants. Commercially available natural inulin mostly comes from chicory
root, dahlia, and Jerusalem artichoke, while synthetic inulin is prepared from sucrose.
Inulin is commonly used in processed foods and as a nondigestible carbohydrate to
replace sweeteners and is utilized in low-calorie foods (Shoaib et al. 2016).
Inulin stimulates the development and metabolic action of a limited number of
bacteria in the colon, particularly Bifidobacteria and Lactobacilli (Kelly 2008).
In vitro trials have demonstrated that colonic fermentation depends on inulin’s
chain length. Fermentation time is dependent on chain length, with longer chain
inulin allowing for the initiation of bacterial fermentation in the distal part of the
colon. The nutritional utilization of inulin and oligofructose provides a possible
way to encourage microbial balance and overcome the difficulties in colonizing
the gut mucosa. For this reason, inulin is used to reduce and replace sugar in food
products. Inulin has been demonstrated to enhance the growth sustainability of
L. acidophilus, L. rhamnosus, and B. lactis when added to milk, yogurt, and ice
cream (Kelly 2009).
The consumption of inulin has demonstrated numerous benefits to the host diges-
tive tract. Inulin has improved bowel movements, GI symptoms, quality of life, and
increased fecal output in various adult populations (Watson et al. 2019). These shifts
are generally attributed to the ability of inulin to reach the colon intact, where inulin
is metabolized by bacteria, leading to beneficial byproducts. These improvements
have generally been associated with modest changes in gut microbiota composition,
specifically Bifidobacteria, Anaerostipes, and Clostridium in constipated subjects
(Vandeputte et al. 2017). A recent randomized, double-blind, placebo-controlled
cross-over trial with inulin isolated chicory root attempted to elucidate the relation-
ship between diet, bowel movement, and colon microbiota. The study demonstrated
that 10 g/d of inulin improved stool frequency and consistency in older adults who
experience constipation (<1 bowel movement/day); however, they could not detect a
shift in gut microbiota (Watson et al. 2019).
Dietary Modulation of the Gut Microbiota by Prebiotics 163
NON-CARBOHYDRATE PREBIOTICS
Conventional dietary knowledge suggests that a diet high in plant material is ben-
eficial for the host’s gut microbiota. As discussed in previous chapters, numerous
studies in humans have examined the dietary impact on gut microbiota composition
and divided the population into two groups: those that eat fruits and vegetables over a
certain level and those that do not. In addition, the relative abundance of one phylum
of bacteria relative to another can predict the risk of obesity (Singh et al. 2017). The
same phyla are also associated with the consumption of fruits and vegetables. The
correlation between health status, diet, and gut composition is putatively linked to
nondigestible carbohydrates. However, some ingredients are excluded when examin-
ing the relationship between certain foods and gut fermentation.
PolyPhenols
Polyphenols represent a new category of potential prebiotics. Until recently, little was
known about the metabolic fate of polyphenols in the intestinal tract. Berries have
been used for centuries as elixirs to help with gastrointestinal symptoms. In humans,
the intestinal absorption of dietary polyphenols is often slow and largely incomplete;
up to 85% of anthocyanins enter the colon intact and can be used as substrates for
microbial metabolism (Kahle et al. 2006). The capability for anaerobic digestion
of berries in gastrointestinal environments is reflected by increased Bifidobacteria
populations and other microbes that catabolize the diverse berry compounds avail-
able, especially polyphenols (Lacombe et al. 2013).
The use of berries for their prebiotic effect has been tested in clinical trials to
improve the clinical picture and the well-being of patients. Promising beneficial
effects demonstrated with berries include positive shifts in gut microbiota com-
position. In vitro human fecal batch cultures have demonstrated the enrichment of
Lactobacillus and Bifidobacteria with 1 g/L of gallic acid and 200 mg/L of antho-
cyanins (Håkansson et al. 2009). These results demonstrate the benefits of dietary
enrichment of berries and its impact on bacterial communities with unique func-
tional repertoires by promoting their growth in competitive environments.
Bioprotective effects of polyphenols and other nutrients observed in vitro are not
entirely transferable to health effects observed in vivo; therefore, it is essential to use
in vivo models to understand mechanisms. Recent studies using the Sprague Dawley
(SD) rat model, fed polyphenols extracted from blueberry and blackberry via gavage,
demonstrated increased Lactobacillus and Bifidobacteria (Bò et al. 2009; Vendrame
et al. 2013). Actinobacteria, with known impacts on human health, namely Slackia,
Bifidobacteria, and Coriobacteriaceae spp, were detected in higher abundance
in SD rats fed a blueberry-enriched diet (Lacombe et al. 2013, 2012). In humans,
dietary treatment with blueberry increased the population of Bifidobacteria more
than two-fold, demonstrating the prebiotic activity of berry polyphenols (Vendrame
et al. 2013). For clinicians, these findings are significant due to the growing interest
in probiotic bacteria and the perceived benefit of increasing their numbers in the GIT
to attain their health benefits.
164 The Gut Microbiome: Bench to Table
volunteers (Guigoz et al. 2002). Notably, infants supplemented with FOS have shown
a reduced risk of immune diseases (Moro et al. 2006). GOS improves the activity
of natural killer cells by increasing the IL-8, IL-10, and C-reactive protein (Vulevic
et al. 2013, 2015). It has also been reported to prevent the risks of atopic dermatitis
and eczema (Moro et al. 2006).
Necrotizing enterocolitis (NEC) is a severe gastrointestinal necrosis condition in
premature neonates. It is one of the most common and often fatal intestinal disor-
ders in preterm infants. Clinical trials have demonstrated a high correlation between
HMO in breast milk and positive outcome measures in NEC progression (Bode
2015; Autran et al. 2018). As previously discussed, HMOs have a bifidogenic effect
in the gut and protect neonates from NEC and pathogenic bacterial infection (Knol
et al. 2005). However, increased levels of bifidogenic oligosaccharides outcompet-
ing pathogens do not entirely describe HMOs’ beneficial effects on NEC (Bode
2015). A randomized study revealed that FOS and GOS enhance the population of
Bifidobacteria but had no significant consequence in controlling the progression
of NEC (Srinivasjois, Rao, & Patole, 2009). Research on HMOs has demonstrated
improved immunity benefits similar to breastfed infants (Bode 2015). Preclinical
research also shows that HMOs reduce intestinal discomfort, reduce food allergy
symptoms, and enhance cognition, leading to myriad health benefits for infants
(Autran et al. 2018). The link between HMO and necrotizing colitis is associated
with competitive binding within the intestinal glycocalyx (Bode 2015). For patho-
gens to survive in the gut, they must attach to epithelial cells; otherwise, they will be
eliminated through excretion. This process occurs through bacterial lectins attach-
ing to glycans on the surface of epithelial cells. In the case of necrotizing colitis,
there is a specific oligosaccharide called disialyllacto-N-tetraose (DSLNT), which
is associated with protective effects in mouse models (Bode 2006). A human cohort
study with preterm infants demonstrated that necrotizing colitis was lower in infants
who received higher amounts of DSLNT from their mothers (Bode 2006). DSLNT
content in breast milk is a potential noninvasive marker to identify infants at risk of
developing NEC and screen high-risk donor milk. In addition, DSLNT could serve
as a natural template to develop novel therapeutics against this devastating disorder.
This association is significant to note when considering therapeutic applications of
HMO in infant formula (Autran et al. 2018).
include improving general body conditions, reducing the risk of specific diseases,
and potential use for the prevention and treatment of illnesses (Bigliardi and Galati
2013). Functional foods differentiate themselves from conventional foods consumed
in a standard diet by containing bioactive components whose physiological activity
is scientifically proven (Bigliardi and Galati 2013). Categories of functional foods
include those that naturally contain one of the bioactive components, fortified foods
in which the native bioactive level is enhanced, and processed foods in which the
bioactive compound is intentionally added (Bigliardi and Galati 2013).
A typical Western diet contains moderate levels of prebiotics through consum-
ing various fruits and vegetables. However, the number of prebiotics contained in
many fruits and vegetables is not sufficient or not consumed in an amount that would
significantly alter the composition of intestinal microflora (Jovanovic-Malinovska,
Kuzmanova, and Winkelhausen 2014). Therefore, consumers look toward supple-
mentation to increase the effective dose of many prebiotics. This affects the puta-
tive mechanism of action by altering the relative abundance of specific nondigestible
components in the diet. The mechanism by which prebiotics can attenuate the pro-
gression of diseases is of great interest to researchers and clinicians.
Fiber supplementation is one of the most common applications for prebiotics in
the Western diet. Most (90%) of the US population does not consume 15 g/d of rec-
ommended dietary fiber doses. Many consumers turn to fiber supplements, typically
isolated from a single food source (McRorie et al. 2015). However, of the fiber supple-
ments on the market today, only a minority possess the physical characteristics that
underlie the mechanisms driving clinically meaningful health benefits. Prebiotics
are clinically accepted and recommended as dietary supplements for the treatment of
constipation. These carbohydrates and prebiotics are fermented in the large intestine
and diminish the transport time of assimilated food by hydrating and increasing acid
production (Den Hond, Geypens, and Ghoos 2000). Intestinal-associated diseases
like IBS and CD are also thought to benefit from these fermentative processes.
Many prebiotic functional food formulations try to take advantage of the inher-
ent presence of probiotics in the food product. Dairy foods are a popular probiotic
food category worldwide and intrinsically contain nutrients that maintain the pro-
biotic organism. Examples of popular probiotic functional foods are yogurt, which
represents about 35% of sales overall, followed by cultured drinks, kefir, cheese,
ice cream, and infant formula (Tamime et al. 2006; Saavedra and Tschernia 2022).
Fermented products, such as yogurt, kefir, and cheese are commonly enhanced pro-
biotic cultures, and more recently, unfermented dairy matrices, such as ice cream
and butter, have also been enhanced with probiotic cultures (Koebnick et al. 2003).
Whey protein, which is rich in nutrients and potential prebiotics, has been forti-
fied with probiotics (Tenorio et al. 2010; Suomalainen et al. 2006). The functional
use of whey, a byproduct of cheesemaking that is often discarded, has been a focus of
functional food development (Tenorio et al. 2010). The commercially available oligo-
saccharides are usually derived from cow milk, plants, and fermented processes and
are very different structurally from what is found in HMO (Bode 2015). Commercial
GOS and FOS tend to be linear carbohydrate chains (Vulevic et al. 2013). However,
little is known about oligosaccharides’ structure–function relationship regarding
therapeutic applications (Vulevic et al. 2015).
168 The Gut Microbiome: Bench to Table
Current knowledge stems from associated genetic differences from cohort studies
and cell/animal models observed within populations. For example, we now know that
HMO is not just a preferential food source for Bifidobacteria but have antimicrobial
properties and can outcompete other bacteria for binding site in the intestinal tract
and directly modulate epithelial cell responses (Bode 2015). Applying HMO formu-
lation in the adult population is much more complicated than in infants. Although
there are many products on the market that claim they can provide increased gut
function, they do not take into account the diversity of HMO (Bode 2015). Adults
are not blank slates when it comes to gut microbiota composition, which means its
alteration is much more difficult. In addition, diet and lifestyle are very heterogone-
ous among humans, generating many variations in the human datasets (Bode 2015).
This variation makes causal relationships between the structure/function of HMO
and the gut microbiota challenging to establish.
Many consumers have allergies to milk proteins, lactose intolerance, and may
have adopted diet choices that partially or entirely exclude food of animal origin.
In response to the increased market for dairy products of nonanimal origin, plant-
based probiotic/prebiotic products are available to consumers. Many raw materials
of plant origin often contain prebiotic substances, which can stimulate the growth
of probiotic microorganisms both during the processing steps and during storage
(Veereman 2007; İspirli, Demirbaş, and Dertli 2018). The use of nuts and grains,
such as almonds, rice, and oats, has been widely adopted as animal milk as these
products are generally perceived as healthy by the consumer and have an appealing
sensory profile (Håkansson et al. 2009; Vulevic et al. 2015). These foods contain
additional functional benefits, such as fibers, vitamins, minerals, flavonoids, and
antioxidants. Enrichment with probiotic cultures is carried out through osmotic
dehydration or vacuum impregnation techniques or by directly adding the micro-
bial cells to juices. A fermentation step may occur in the latter case, creating addi-
tional bioactive nutrients.
conditions of the host gut. Intrinsic features of the food matrix can strongly endanger
their viability in food, processing treatments applied, competitive background flora,
conditions, and duration of storage. Probiotic microorganisms are relatively sensitive
compared to some spoilage microorganisms, and the number of viable bacteria can
decrease by 1–2 log CFU (colony-forming unit)/g or even more during production
and storage. Minimum values of at least 106 –107 CFU/g are needed for residency
in the gut; therefore, the microbial dose in the original product needs to be 108–109
probiotic cells (Jayamanne and Adams 2006). Since the health effects depend on the
conditions of the product at the time of consumption, the maintenance of microbial
viability must be ensured over its entire shelf-life.
The carrier matrix has a significant effect on the quality of probiotic products, as its
composition and characteristics can alter the viability and effectiveness of probiot-
ics. One of the main obstacles is represented by the acid pH since Lactobacilli have
an optimal growth pH of 5.5–6.0 and Bifidobacteria of 6.0–7.0 (De Vuyst, 2000).
Acidic pH, which is relevant for fermented foods, fruit, and vegetables, leads to an
increase in the concentration of organic acids in the undissociated form, therefore
increasing the antimicrobial effect of these acids. Furthermore, at low pH values
(below 4.5), cells need significant energy to keep the intracellular pH constant, seiz-
ing adenosine triphosphate from other cellular needs, causing cell death. Low pH
(approx. 3.5) was shown to be the main culprit in the loss of viability of L. rhamno-
sus, L. plantarum, and L. casei in cherry juice, especially during refrigerated storage
(Nematollahi et al. 2016). In fermented milk, L. acidophilus tolerates acid conditions
better than Bifidobacteria, proving the careful selection of the strains used in each
probiotic food to be crucial (Tamime et al. 2006).
Water content in food formulations, whether expressed as moisture content or water
activity (aw), plays a determining role in the survival of probiotics in food. The aw, i.e.,
the availability of water for microbial growth, varies based on the product’s intended
shelf. If a food product has a water activity range above 0.85, it will have to be refrig-
erated or use another barrier to control pathogen growth (Gomand et al. 2019). At aw
above 0.85, there is a substantial loss of prebiotic viability as other spoilage microor-
ganisms compete for nutrients. In contrast, a food product with a water activity between
0.60 and 0.85 will not require refrigeration but will have a limited shelf-life due to
yeasts and molds. Lastly, food with a water activity below 0.60 will have an extended
shelf-life, even without refrigeration. With probiotic formulations, food manufacturers
can employ several tactics, including drying, freezing, or adding solutes like salt or
sugar, helping to reduce their products’ water activity levels, extending their shelf-life.
There is a potential benefit to adding probiotics to low moisture or intermedi-
ate moisture foods, such as chocolate, peanut butter, cereals, and dried fruit paste
because they are stable for prolonged periods (Finn et al. 2013). For example, a via-
bility loss of less than 1 log CFU/g of L. rhamnosus was observed in peanut butter
(aw approx. 0.35) stored at 25°C for 27 weeks (Klu et al. 2012). In addition, osmolarity
in foods containing high amounts of sugar or salt, such as fruit purees, jams, or aged
cheeses, plays a role in the survival of microorganisms in foods. Generally, sugar and
Dietary Modulation of the Gut Microbiota by Prebiotics 171
salt greatly influence the growth and survival of probiotics in food as their presence
increases osmotic pressure. Salt is widely used in foods to prolong its shelf-life, but
at high concentrations (~4%–6%), it can put the viability of probiotics at risk. The
tolerance to osmotic stress is strain-specific and depends on the phospholipidic com-
position of the membrane. Lactobacillus casei demonstrated slower growth in the
presence of high glucose concentrations, as well as Bifidobacteria, while L. planta-
rum demonstrated more resistance to osmotic stress (Homayouni et al. 2008).
Since most probiotic species are strictly anaerobic, dissolved oxygen in the matrix
is critical for their survival in foods, as hydrogen peroxide is formed through differ-
ent mechanisms both at intracellular and extracellular levels. Probiotic bacteria are
practically devoid of an oxygen scavenging system, which causes the reduction of
oxygen to hydrogen peroxide. The toxicity of hydrogen peroxide is due to its ability
to block fructose-6-phosphofructoketolase, a key enzyme for sugar metabolism. In
addition, superoxide and hydroxyl radicals are formed in dissolved oxygen due to the
oxidation of membrane lipids in the cell, which induces DNA damage and causes cell
death (Önneby et al. 2013). Different processing steps can contribute to the enrich-
ment of the matrix in dissolved oxygen, such as homogenization, cooling after heat
treatment, and mixing with ingredients. Although all probiotics are anaerobic, oxy-
gen tolerance varies enormously between genera and species. Indeed, Bifidobacteria
tend to be more sensitive than lactic acid bacteria. In the presence of oxygen, the most
sensitive bacteria change the profile of membrane fatty acids and show an elongation
of the lag phase and a substantial reduction in lactate produced (Looijer-van Langen
and Dieleman 2009; Patel and Denning 2013).
Probiotic microorganisms can be exposed to thermal stress when producing food-
containing probiotics. When heat treatments are used to inactivate pathogenic and
spoiler microorganisms in raw materials, thermal stress on probiotics can be easily
avoided by inoculating them downstream of the heat treatment or by using probiotic
species other than lactic acid bacteria and Bifidobacteria. The genus Bacillus coagulans
has been successfully used in the processing of various foods such as banana muffins
and waffles, brewed coffee, chocolate fudge frosting, and hot fudge topping, and the loss
of viability has been less than 1 log CFU/g in all cases (Majeed et al. 2016). However,
during fermentation, probiotics may undergo thermal stress. The production of yogurt
often requires that the milk is held at temperatures above 40°C–42°C, which can be
harmful to probiotics. Therefore, the industry has curated cultivars of probiotics more
resistant to thermal treatment. In yogurt making, the incubation temperature can be as
high as 44°C, which causes a loss in viability of L. acidophilus and Bifidobacterium
BB-12. At higher fermentation temperatures (e.g., 45°C), more heat-resistant L. brueckii
subsp. bulgaricus becomes the dominant species and produces a large amount of lactic
acid, hydrogen peroxide, and sometimes bacteriocins, which causes probiotic growth to
stop (Nematollahi et al. 2016). Therefore, to foster the biodiversity of probiotics, some
manufacturers inoculate species after the fermentation process.
Freezing is a critical process in the manufacture of ice cream. However, it presents
a technical challenge in the development of probiotic-enhancing products. Subzero
temperatures are detrimental to microbial viability because as the portion of fro-
zen water increases, the aw value of the liquid solution decreases. Furthermore,
osmotic pressure increases as water diffuses outside the cell, dehydrates, and freezes
172 The Gut Microbiome: Bench to Table
the extracellular water. The mechanical stress due to ice crystals damages the mem-
branes, the low temperatures shock the cells, the (harmful) solutes concentrate, and
the cells become dehydrated. Together with the progressive lowering of the tempera-
ture, all this determines the so-called cold shock, which causes cell death. In probi-
otic ice cream, L. rhamnosus loses viability during freezing of about 1.8 log CFU/g,
probably due to a combined effect of temperature lowering and oxygen incorporation
during mixing (Marino et al. 2017).
Developers of enhanced probiotic products attempt to harness the intrinsic pro-
tective substances that protect the viability of probiotics from the stresses accumu-
lated in the food matrix and during the production process and the GIT transit. The
compounds most frequently used for this purpose are carbohydrates, and many have
additional prebiotic effects. Sugars, especially those with low molecular weight,
interact with the polar heads in the cell membranes, replacing the water molecules
and stabilizing the membranes themselves during osmotic stresses. Low-molecular-
weight sugars, such as trehalose, sucrose, and lactulose, can improve the survival of
probiotics during freezing, dehydration, and storage both in products of animal and
plant origin (Betoret et al. 2017). Besides, they have also been shown to have direct
functional activities, such as a bifidogenic effect and an increase in the hydrophobic-
ity of the surface of probiotic cells, which is related to the ability to adhere to and
interact with the intestinal wall (Dlamini et al. 2019).
Even the most complex polysaccharides and fibers can improve the viability of
probiotics in food. These are dietary compounds that reach the gut in an undigested
form and are selectively fermented by the intestinal microbiota, thus stimulating
their growth and activity. However, their stimulation activity also aids the probiot-
ics added to foods. Consequently, the development of functional foods that contain
both probiotics and prebiotics (symbiotic foods) can provide a double physiologi-
cal advantage. Typically, prebiotic substances mainly come from plant food sources.
Inulin and FOS, widely used for this purpose, are commonly extracted from onions,
bananas, wheat, artichokes, garlic, and other whole foods. Other widely used oli-
gosaccharides, such as GOS and HMO, are of animal origin (Anadón, Martínez-
Larrañaga, Arés, & Martínez, 2016).
Even microorganisms are capable of producing extracellular substances that act
as prebiotics, such as exopolysaccharide (İspirli, Demirbaş, and Dertli 2018). The
effectiveness of prebiotics in the context of functional probiotic foods is linked to
the fact that these substances can be metabolized and used as a source of energy
by probiotics. Furthermore, owing to their physicochemical characteristics, they can
effectively protect cell envelopes, increase the glass transition temperatures in the
aqueous phase, retain water, and prevent the formation of ice crystals that may cause
mechanical damage to the cells themselves (Tymczyszyn et al. 2011). Due to these
activities, the effects essentially protect probiotics against acidity, high temperatures,
and dehydration. Their addition to food improves the stability of probiotics during
processing, storage, and in vitro digestion. In the blended carrot and orange juices,
the presence of 2% inulin improved the survival of L. plantarum during storage for
30 days at 4°C and during in vitro digestion, and a mixture of inulin, FOS, and GOS
had a similar effect on L. casei in a blended red fruit beverage (Bernal-Castro, Díaz-
Moreno, and Gutiérrez-Cortés 2019).
Dietary Modulation of the Gut Microbiota by Prebiotics 173
CONCLUSION
As the concept of prebiotics continues to evolve, researchers investigate the potential
benefits of their ingestion. There are many possible sources for prebiotics, which can
be found in the natural environment or be synthesized based on research indicating
certain functional groups with benefits. However, more work is needed to determine
which prebiotic regimen is best for dietary recommendations, while recommenda-
tions cannot probably be applied ubiquitously across the population. The future of
prebiotic supplementations may rely upon the intended hosts existing microbiome
and genetic profile. This opens a fascinating field of inquiry into tailoring probiotic
and prebiotic regimens for optimum health.
REFERENCES
Addison, Ruth, Lauren Hill, Lars Bode, Bianca Robertson, Biswa Choudhury, David
Young, Charlotte Wright, Clare Relton, Ada L. Garcia, and David M. Tappin. 2019.
“Development of a Biochemical Marker to Detect Current Breast Milk Intake.”
Maternal and Child Nutrition, no. April 2019: 1–8. https://doi.org/10.1111/mcn.12859.
Autran, Chloe A., Benjamin P. Kellman, Jae H. Kim, Elizabeth Asztalos, Arlin B. Blood, Erin C.
Hamilton Spence, Aloka L. Patel, Jiayi Hou, Nathan E. Lewis, and Lars Bode. 2018.
“Human Milk Oligosaccharide Composition Predicts Risk of Necrotising Enterocolitis
in Preterm Infants.” Gut 67 (6): 1064–70. https://doi.org/10.1136/GUTJNL-2016-312819.
Azad, Meghan B., Bianca Robertson, Faisal Atakora, Allan B. Becker, Padmaja Subbarao,
Theo J. Moraes, Piushkumar J. Mandhane, et al. 2018. “Human Milk Oligosaccharide
Concentrations Are Associated with Multiple Fixed and Modifiable Maternal
Characteristics, Environmental Factors, and Feeding Practices.” Journal of Nutrition
148 (11): 1733–42. https://doi.org/10.1093/jn/nxy175.
Bernal-Castro, Camila Andrea, Consuelo Díaz-Moreno, and Carolina Gutiérrez-Cortés.
2019. “Inclusion of Prebiotics on the Viability of a Commercial Lactobacillus Casei
Subsp. Rhamnosus Culture in a Tropical Fruit Beverage.” Journal of Food Science and
Technology 56 (2): 987. https://doi.org/10.1007/S13197-018-03565-W.
Betoret, E., L. Calabuig-Jiménez, F. Patrignani, R. Lanciotti, and M. Dalla Rosa. 2017.
“Effect of High Pressure Processing and Trehalose Addition on Functional Properties
of Mandarin Juice Enriched with Probiotic Microorganisms.” LWT – Food Science and
Technology 85 (November): 418–22. https://doi.org/10.1016/J.LWT.2016.10.036.
Bigliardi, Barbara, and Francesco Galati. 2013. “Innovation Trends in the Food Industry:
The Case of Functional Foods.” Trends in Food Science and Technology 31 (2): 118–29.
https://doi.org/10.1016/J.TIFS.2013.03.006.
Bode, Lars. 2006. “Recent Advances on Structure, Metabolism, and Function of Human Milk
Oligosaccharides.” The Journal of Nutrition. https://doi.org/10.1093/jn/136.8.2127.
———. 2009. “Human Milk Oligosaccharides: Prebiotics and Beyond.” Nutrition Reviews.
https://doi.org/10.1111/j.1753-4887.2009.00239.x.
———. 2012. “Human Milk Oligosaccharides: Every Baby Needs a Sugar Mama.”
Glycobiology 22 (9): 1147–62. https://doi.org/10.1093/glycob/cws074.
———. 2015. “The Functional Biology of Human Milk Oligosaccharides.” Early Human
Development. https://doi.org/10.1016/j.earlhumdev.2015.09.001.
———. 2018. “Human Milk Oligosaccharides in the Prevention of Necrotizing Enterocolitis:
A Journey from In Vitro and In Vivo Models to Mother-Infant Cohort Studies.”
Frontiers in Pediatrics. https://doi.org/10.3389/fped.2018.00385.
Bode, Lars, and Evelyn Jantscher-Krenn. 2012. “Structure-Function Relationships of Human
Milk Oligosaccharides.” Advances in Nutrition. https://doi.org/10.3945/an.111.001404.
174 The Gut Microbiome: Bench to Table
Cavaglieri, Claudia R., Anita Nishiyama, Luis Claudio Fernandes, Rui Curi, Elizabeth A. Miles,
and Philip C. Calder. 2003. “Differential Effects of Short-Chain Fatty Acids on Proliferation
and Production of Pro- and Anti-Inflammatory Cytokines by Cultured Lymphocytes.”
Life Sciences 73 (13): 1683–90. https://doi.org/10.1016/S0024-3205(03)00490-9.
Cheplin, Harry Asber, and Leo Frederick Rettger. 2011. A Treatise on the Transformation
of the Intestinal Flora, with Special Reference to the Implantation of Bacillus
Acidophilus. A Treatise on the Transformation of the Intestinal Flora, with Special
Reference to the Implantation of Bacillus Acidophilus. https://doi.org/10.5962/bhl.
title.24080.
Claus, Sandrine P., Sandrine L. Ellero, Bernard Berger, Lutz Krause, Anne Bruttin, Jérôme
Molina, Alain Paris, et al. 2011. “Colonization-Induced Host-Gut Microbial Metabolic
Interaction.” MBio 2 (2). https://doi.org/10.1128/mBio.00271-10.
Collins, Stephanie, and Gregor Reid. 2016. “Distant Site Effects of Ingested Prebiotics.”
Nutrients 8 (9): 523. https://doi.org/10.3390/nu8090523.
Danneskiold-Samsøe, Niels Banhos, Helena Dias de Freitas Queiroz Barros, Rosangela
Santos, Juliano Lemos Bicas, Cinthia Baú Betim Cazarin, Lise Madsen, Karsten
Kristiansen, Glaucia Maria Pastore, Susanne Brix, and Mário Roberto Maróstica
Júnior. 2019. “Interplay between Food and Gut Microbiota in Health and Disease.”
Food Research International. https://doi.org/10.1016/j.foodres.2018.07.043.
David, Lawrence A., Corinne F. Maurice, Rachel N. Carmody, David B. Gootenberg, Julie E.
Button, Benjamin E. Wolfe, Alisha V. Ling, et al. 2014. "Diet Rapidly and Reproducibly
Alters the Human Gut Microbiome." Nature 505 (7484): 559–63. https://doi.org/10.1038/
nature12820.
Del Bo’, Cristian, Salvatore Ciappellano, Dorothy Klimis-Zacas, Daniela Martini, Claudio
Gardana, Patrizia Riso, and Marisa Porrini. 2009. “Anthocyanin Absorption, Metabolism,
and Distribution from a Wild Blueberry-Enriched Diet (Vaccinium Angustifolium) Is
Affected by Diet Duration in the Sprague−Dawley Rat.” Journal of Agricultural and
Food Chemistry 58 (4): 2491–97. https://doi.org/10.1021/JF903472X.
Del Bo’, Cristian, Daniela Martini, Stefano Vendrame, Patrizia Riso, Salvatore Ciappellano,
Dorothy Klimis-Zacas, and Marisa Porrini. 2010. “Improvement of Lymphocyte
Resistance against H2O2-Induced DNA Damage in Sprague-Dawley Rats after Eight
Weeks of a Wild Blueberry (Vaccinium Angustifolium)-Enriched Diet.” Mutation
Research – Genetic Toxicology and Environmental Mutagenesis 703 (2): 158–62.
https://doi.org/10.1016/j.mrgentox.2010.08.013.
Di Criscio, T., A. Fratianni, R. Mignogna, L. Cinquanta, R. Coppola, E. Sorrentino, and G. Panfili.
2010. “Production of Functional Probiotic, Prebiotic, and Synbiotic Ice Creams.” Journal
of Dairy Science 93 (10): 4555–64. https://doi.org/10.3168/JDS.2010-3355.
Dlamini, Ziyanda C., Rashwahla L.S. Langa, Olayinka A. Aiyegoro, and Anthony I. Okoh.
2019. “Safety Evaluation and Colonisation Abilities of Four Lactic Acid Bacteria as
Future Probiotics.” Probiotics and Antimicrobial Proteins 11 (2): 397–402. https://doi.
org/10.1007/S12602-018-9430-Y.
Elizabeth Tymczyszyn, E., Esteban Gerbino, Andrés Illanes, and Andrea Gómez-Zavaglia.
2011. “Galacto-Oligosaccharides as Protective Molecules in the Preservation of
Lactobacillus Delbrueckii Subsp. Bulgaricus.” Cryobiology 62 (2): 123–29. https://doi.
org/10.1016/J.CRYOBIOL.2011.01.013.
Fanaro, Silvia, Günther Boehm, Johan Garssen, Jan Knol, Fabio Mosca, Bernd Stahl, and
Vittorio Vigi. 2005. “Galacto-Oligosaccharides and Long-Chain Fructo-Oligosaccharides
as Prebiotics in Infant Formulas: A Review.” Acta Paediatrica, International Journal of
Paediatrics, Supplement 94 (449): 22–26. https://doi.org/10.1080/08035320510043538.
Fernandes, Ricardo, Vinicius A. do Rosario, Michel C. Mocellin, Marilyn G.F. Kuntz,
and Erasmo B.S.M. Trindade. 2017. “Effects of Inulin-Type Fructans, Galacto-
Oligosaccharides and Related Synbiotics on Inflammatory Markers in Adult Patients
Dietary Modulation of the Gut Microbiota by Prebiotics 175
İspirli, Hümeyra, Fatmanur Demirbaş, and Enes Dertli. 2018. “Glucan Type Exopolysaccharide
(EPS) Shows Prebiotic Effect and Reduces Syneresis in Chocolate Pudding.” Journal of
Food Science and Technology 55 (9): 3821–26. https://doi.org/10.1007/S13197-018-3181-3.
Jayamanne,Vijith, and Steve Munyard Adams. 2006. “Determination of Survival, Identity
and Stress Resistance of Probiotic Bifidobacteria in Bio-Yoghurts.” Letters in Applied
Microbiology 42 (3): 189–94. https://doi.org/10.1111/J.1472-765X.2006.01843.X.
Jovanovic-Malinovska, Ruzica, Slobodanka Kuzmanova, and Eleonora Winkelhausen.
2014. “Oligosaccharide Profile in Fruits and Vegetables as Sources of Prebiotics and
Functional Foods.” 17 (5): 949–65. https://doi.org/10.1080/10942912.2012.680221.
Kahle, Kathrin, Michael Kraus, Wolfgang Scheppach, Matthias Ackermann, Friederike
Ridder, and Elke Richling. 2006. “Studies on Apple and Blueberry Fruit Constituents:
Do the Polyphenols Reach the Colon after Ingestion?” Molecular Nutrition and Food
Research 50: 418–23. https://doi.org/10.1002/mnfr.200500211.
Keenan, Derek F., Nigel Brunton, Francis Butler, Rudy Wouters, and Ronan Gormley. 2011.
“Evaluation of Thermal and High Hydrostatic Pressure Processed Apple Purees Enriched
with Prebiotic Inclusions.” Innovative Food Science and Emerging Technologies 12 (3):
261–68. https://doi.org/10.1016/J.IFSET.2011.04.003.
Kelly, Greg. 2008. “Inulin-Type Prebiotics – A Review: Part 1.” Alternative Medicine Review:
A Journal of Clinical Therapeutic 13 (4): 315–29.
———. 2009. “Inulin-Type Prebiotics: A Review (Part 2).” Alternative Medicine Review.
https://doi.org/10.1519/SSC.0b013e318281f689.
Khangwal, Ishu, and Pratyoosh Shukla. 2019. “Potential Prebiotics and Their Transmission
Mechanisms: Recent Approaches.” Journal of Food and Drug Analysis 27 (3): 649–56.
https://doi.org/10.1016/J.JFDA.2019.02.003.
Klu, Yaa Asantewaa Kafui, Jonathan H. Williams, Robert D. Phillips, and Jinru Chen.
2012. “Survival of Lactobacillus Rhamnosus GG as Influenced by Storage Conditions
and Product Matrixes.” Journal of Food Science 77 (12). https://doi.org/10.1111/
J.1750-3841.2012.02969.X.
Knol, Jan, Petra Scholtens, Corinna Kafka, Jochem Steenbakkers, Sabine Groß, Klaus Helm,
Malte Klarczyk, Helmut Schöpfer, Heinz Michael Böckler, and John Wells. 2005.
“Colon Microflora in Infants Fed Formula with Galacto- and Fructo-Oligosaccharides:
More like Breast-Fed Infants.” Journal of Pediatric Gastroenterology and Nutrition 40
(1): 36–42. https://doi.org/10.1097/00005176-200501000-00007.
Kober, Lars, Christoph Zehe, and Juergen Bode. 2013. “Optimized Signal Peptides for the
Development of High Expressing CHO Cell Lines.” Biotechnology and Bioengineering.
https://doi.org/10.1002/bit.24776.
Koebnick, Corinna, Irmtrud Wagner, Peter Leitzmann, Udo Stern, and H. J.Franz Zunft.
2003. “Probiotic Beverage Containing Lactobacillus Casei Shirota Improves
Gastrointestinal Symptoms in Patients with Chronic Constipation.” Canadian Journal
of Gastroenterology = Journal Canadien de Gastroenterologie 17 (11): 655–59. https://
doi.org/10.1155/2003/654907.
Lacombe, Alison, Robert. Li, Dorothy Klimis-Zacas, ALekzandra Kristo, Shravani Tadepalli,
Emily Krauss, Ryan Young, and Vivian C.H.. Wu. 2012. “Lowbush Blueberries,
Vaccinium Angustifolium, Modulate the Functional Potential of Nutrient Utilization
and DNA Maintenance Mechanisms in the Rat Proximal Colon Microbiota.” Functional
Foods in Health and Disease 2 (6). https://doi.org/10.31989/ffhd.v2i6.87.
———. 2013. “Lowbush Wild Blueberries Have the Potential to Modify Gut Microbiota and
Xenobiotic Metabolism in the Rat Colon.” PLoS One 8 (6). https://doi.org/10.1371/jour-
nal.pone.0067497.
Langkamp-Henken, Bobbi, Bradley S. Bender, Elizabeth M. Gardner, Kelli A. Herrlinger-
Garcia, Michael J. Kelley, Donna M. Murasko, Joseph P. Schaller, Joyce K. Stechmiller,
Debra J. Thomas, and Steven M. Wood. 2004. “Nutritional Formula Enhanced Immune
Dietary Modulation of the Gut Microbiota by Prebiotics 177
Revolledo, Lillana., C. S.A. Ferreira, and Antonio J.P. Ferreira. 2009. “Prevention of Salmonella
Typhimurium Colonization and Organ Invasion by Combination Treatment in Broiler
Chicks.” Poultry Science 88 (4): 734–43. https://doi.org/10.3382/PS.2008–00410.
Saavedra, Lucas., and A. Tschernia. 2022. “Human Studies with Probiotics and Prebiotics:
Clinical Implications.” https://doi.org/10.1079/BJN/2002543.
Scott, Karen P., Jenny C. Martin, Christophe Chassard, Marlene Clerget, Joanna Potrykus,
Gill Campbell, Claus Dieter Mayer, et al. 2011. “Substrate-Driven Gene Expression in
Roseburia Inulinivorans: Importance of Inducible Enzymes in the Utilization of Inulin
and Starch.” Proceedings of the National Academy of Sciences of the United States of
America 108 (Suppl. 1): 4672–79. https://doi.org/10.1073/pnas.1000091107.
Shoaib, Muhammad, Aamir Shehzad, Mukama Omar, Allah Rakha, Husnain Raza,
Hafiz Rizwan Sharif, Azam Shakeel, Anum Ansari, and Sobia Niazi. 2016. “Inulin:
Properties, Health Benefits and Food Applications.” Carbohydrate Polymers. https://
doi.org/10.1016/j.carbpol.2016.04.020.
Singh, Rasnik K., Hsin Wen Chang, Di Yan, Kristina M. Lee, Derya Ucmak, Kirsten Wong,
Michael Abrouk, et al. 2017. “Influence of Diet on the Gut Microbiome and Implications
for Human Health.” Journal of Translational Medicine 15 (1): 1–17. https://doi.org/
10.1186/S12967-017-1175-Y.
Tamime, AdnanY., Maria Saarela, A. Korslund Søndergaard, V.V. Mistry, and N.P. Shah.
2006. “Production and Maintenance of Viability of Probiotic Micro-Organisms in
Dairy Products.” Probiotic Dairy Products (November): 39–72. https://doi.org/10.1002/
9780470995785.CH3.
Tenorio, María Dolores, Irene Espinosa-Martos, Guadalupe Préstamo, and Pilar Rupérez. 2010.
“Soybean Whey Enhance Mineral Balance and Caecal Fermentation in Rats.” European
Journal of Nutrition 49 (3): 155–63. https://doi.org/10.1007/S00394-009-0060-8.
Vandeputte, Doris, Gwen Falony, Sara Vieira-Silva, Jun Wang, Manuela Sailer, Stephan
Theis, Kristin Verbeke, and Jeroen Raes. 2017. “Prebiotic Inulin-Type Fructans Induce
Specific Changes in the Human Gut Microbiota.” Gut 66 (11): 1968–74. https://doi.
org/10.1136/gutjnl-2016-313271.
Veereman, Gigi. 2007. “Pediatric Applications of Inulin and Oligofructose.” The Journal of
Nutrition 137 (11): 2585S–2589S. https://doi.org/10.1093/JN/137.11.2585S.
Vendrame, Stefano, Allison Daugherty, Aleksandra S. Kristo, Patrizia Riso, and Dorothy
Klimis-Zacas. 2013. “Wild Blueberry (Vaccinium Angustifolium) Consumption Improves
Inflammatory Status in the Obese Zucker Rat Model of the Metabolic Syndrome.” Journal
of Nutritional Biochemistry 24 (8): 1508–12. https://doi.org/10.1016/j.jnutbio.2012.12.010.
Vendrame, Stefano, Simone Guglielmetti, Patrizia Riso, Stefania Arioli, Dorothy Klimis-
Zacas, and Marisa Porrini. 2011. “Six-Week Consumption of a Wild Blueberry Powder
Drink Increases Bifidobacteria in the Human Gut.” Journal of Agricultural and Food
Chemistry 59 (24): 12815–20. https://doi.org/10.1021/jf2028686.
Vulevic, Jelena, Aleksandra Juric, George Tzortzis, and Glenn R. Gibson. 2013. “A Mixture
of Trans-Galactooligosaccharides Reduces Markers of Metabolic Syndrome and
Modulates the Fecal Microbiota and Immune Function of Overweight Adults 1–3.”
Journal of Nutrition 143 (3): 324–31. https://doi.org/10.3945/jn.112.166132.
Vulevic, Jelena, Aleksandra Juric, Gemma E. Walton, Sandrine P. Claus, George Tzortzis,
Ruth E. Toward, and Glenn R. Gibson. 2015. “Influence of Galacto-Oligosaccharide
Mixture (B-GOS) on Gut Microbiota, Immune Parameters and Metabonomics in Elderly
Persons.” The British Journal of Nutrition 114 (4): 586–95. https://doi.org/10.1017/
S0007114515001889.
Wang, Shumin, Yue Xiao, Fengwei Tian, Jianxin Zhao, Hao Zhang, Qixiao Zhai, and Wei
Chen. 2020a. “Rational Use of Prebiotics for Gut Microbiota Alterations: Specific
Bacterial Phylotypes and Related Mechanisms.” Journal of Functional Foods 66
(September 2019). https://doi.org/10.1016/j.jff.2020.103838.
Dietary Modulation of the Gut Microbiota by Prebiotics 179
———. 2020b. “Rational Use of Prebiotics for Gut Microbiota Alterations: Specific Bacterial
Phylotypes and Related Mechanisms.” Journal of Functional Foods. Elsevier Ltd.
https://doi.org/10.1016/j.jff.2020.103838.
Watson, Anthony W., David Houghton, Peter J. Avery, Christopher Stewart, Elaine E.
Vaughan, P. Diederick Meyer, Minse J.J. de Bos Kuil, Peter J.M. Weijs, and Kirsten
Brandt. 2019. “Changes in Stool Frequency Following Chicory Inulin Consumption,
and Effects on Stool Consistency, Quality of Life and Composition of Gut Microbiota.”
Food Hydrocolloids. https://doi.org/10.1016/j.foodhyd.2019.06.006.
Wu, Xianli, Liwei Gu, Ronald L. Prior, and Steve McKay. 2004. “Characterization of
Anthocyanins and Proanthocyanidins in Some Cultivars of Ribes, Aronia, and Sambucus
and Their Antioxidant Capacity.” Journal of Agricultural and Food Chemistry 52 (26):
7846–56. https://doi.org/10.1021/JF0486850.
Younes, Hassan, Charles Coudray, Jacques Bellanger, Christian Demigné, Yves Rayssiguier,
and Christian Rémésy. 2001. “Effects of Two Fermentable Carbohydrates (Inulin and
Resistant Starch) and Their Combination on Calcium and Magnesium Balance in Rats.”
British Journal of Nutrition 86 (4): 479–85. https://doi.org/10.1079/bjn2001430.
Section III
T2–3 Effectiveness Trials
in General Population and
Regulatory Limitations
7 Gut Microbiota and
Polyphenols: Discussing
a Powerful Interplay and
Its Effect on Health
Aleksandra S. Kristo
California Polytechnic State University
Dorothy Klimis-Zacas
University of Maine
CONTENTS
Introduction ............................................................................................................ 183
Overview of Polyphenols ....................................................................................... 185
Classification and General Description ............................................................. 185
Food Sources and Consumption........................................................................ 186
Overall Metabolism/Bioavailability and Excretion ............................................... 187
Microbiota Metabolism of Polyphenols and Health Effects .................................. 190
Gut-Derived Metabolites ................................................................................... 190
Health Effects of Polyphenol/Metabolite-Microbiota Interaction .................... 193
Considerations on Limitations of Human Studies, Novel Metabolic
Approaches, and Metabotypes .......................................................................... 201
References ..............................................................................................................205
INTRODUCTION
Gut microbiota and polyphenols, each in their own right, constitute research areas
of increasing interest due to accumulating scientific evidence indicating their sig-
nificant potential benefits and functions in human health and longevity. While
polyphenols have been investigated separately as bioactive compounds that can
confer notable health benefits in an array of metabolic conditions, the association
and interaction with the gut microbiome is less studied while is gaining increasing
interest lately.
Polyphenols, as plant-secondary metabolites minimally processed by human
metabolism, rely heavily on the metabolic capacities of the gut microbiota for
their absorption and potential biological actions. By producing bioactive poly-
phenol-derived compounds through enzymatic activities of a diverse community
items such as apples, berries, citrus fruits, plums, beans, nuts, cocoa, tea, coffee, and
wine are considered good sources of polyphenols and have been to a greater or lesser
degree documented to confer health benefits related to chronic conditions including
cardiovascular disease (CVD) and type 2 diabetes mellitus (T2DM) (Williamson,
2017), along with neurodegenerative diseases (Renaud and Martinoli, 2019). While
polyphenols gained early recognition for their antioxidant capacity, mechanistic
studies employing individual compounds are increasingly documenting their signal-
ing properties via multiple cell signaling mechanisms, commonly related to oxidative
or inflammatory status (Vendrame et al., 2015).
Recently, more emphasis has been placed on the potential interplay of polyphe-
nols and the microbiome. More specifically, work is being conducted to investigate
mechanisms of action involving microbiota-derived metabolites, as these relate to
polyphenol action. The gut microbiome can convert polyphenols into secondary
compounds that may extend health benefits.
Notably, the global polyphenol market, which includes a variety of their uses in
foods, beverages, pharmaceuticals, and cosmetics, was estimated to have exceeded
700 million USD in 2015 and is projected to reach 1.1 billion USD by 2022.
Interestingly, however, there is currently no uniform regulatory framework or rec-
ommendation consensus regarding polyphenol consumption and/or supplementation
or fortification in functional foods (Cory et al., 2018).
OVERVIEW OF POLYPHENOLS
C
TABLE 7.1
Dietary Polyphenols: Their Characteristic Bioactive Compounds and Typical
Food Sources
Dietary
Polyphenol Type Characteristic Bioactive Compound(s)a Dietary Source(s)
Phenolic acids Chlorogenic, caffeic, gallic, ferulic acids Coffee, berries, kiwi, apple, cherry
Phenolic alcohols Hydroxytyrosol Olive
Stilbenes Resveratrol Grapes, red wine (primarily)
Lignans Secolariciresinol Linseed
Flavonoids Genistein, daidzein (isoflavones), Soy, miso
Luteolin, apigenin (flavones) Celery, parsley
Hesperetin, naringenin (flavanones) Oranges, lemon
Quercetin, kaempferol, myricetin (flavonols) Onion, leek, broccoli
(Epi)catechins (flavanols) Grapes, wine, cocoa, apricots
(Epi)gallocatechins (flavanols) Beans, green tea
Epigallocatechin gallate (flavanols) Berries, aubergine
Delphinidin, cyanidin, malvidin
(anthocyanins)
Tannins Procyanidins (condensed tannins) Cocoa, chocolate, apples, grapes
Gallotannins, ellagitannins (hydrolyzable Mango, pomegranate
tannins)
and cereals, while the total intake was estimated to be 1 g/d. Nonetheless, significant
uncertainties were due to the lack of accurate and comprehensive data on the content
of some main polyphenol classes in foods (Scalbert and Williamson, 2000). More
recently, the estimated intake of polyphenols through dietary sources and herbal
medicines and/or supplements combined was at 2 g/d, a higher amount than initially
anticipated (Espin et al., 2017). The types of dietary polyphenols, with characteristic
bioactive compounds and typical dietary sources, can be viewed in Table 7.1, while
the classification of compounds can be seen in Figure 7.1.
Bioactive
compounds
D
i
e
t
a
r
y
S
o
u
r
c
e
s
FIGURE 7.1 Main bioactive compounds as they per tain to polyphenols and respective dieta r y sources displayed (Modified from: K r isto et al., 2016).
The Gut Microbiome: Bench to Table
Powerful Interplay of Gut Microbiota and Polyphenols 189
low, medium, and high responders to nutrient supplementation either through the diet
or other types of oral supplementation (Tressera-Rimbau et al., 2018). Interestingly,
a series of pharmacogenomics studies have demonstrated that for specific drugs,
including some phytochemicals of the alkaloid family like codeine, individuals can
be categorized into poor, intermediate, or extensive, and ultra-rapid metabolizers,
and dosing must be adapted to achieve equivalent plasma or tissue concentration of
the active metabolite (Filipski et al., 2016).
An interesting example of the approach toward a better understanding of the role
of genetic polymorphisms in the variability of the response to diet can be seen in
the case of the Mediterranean diet (MedDiet). More specifically, the benefit of the
Mediterranean diet on cardiovascular‐related outcomes has been shown to depend
significantly on genetic variants of the transcription factor 7‐like 2 (TCF7L2) gene
(Corella et al., 2013). This specific gene encodes for a protein functioning as a tran-
scription factor, and the TCF7L2‐rs7903146 polymorphism is known as a strong
genetic determinant of T2DM risk and fasting plasma glucose concentration regula-
tion (Palmer et al., 2011). More specifically, the prevalence of the 7903146T allele is
associated with higher T2DM risk. In an elegant study, Corella et al. demonstrated
that intervention with the MedDiet was more beneficial toward significantly attenuat-
ing cardiovascular risk factors and stroke incidence for individuals with the 7903146T
allele (Corella et al., 2013). Nonetheless, it must be underlined that the majority of
nutrigenetics studies thus far are observational and association studies, while obtained
results have not been fully confirmed in follow-up intervention experimental studies.
The biological properties of dietary polyphenols as they relate to health benefits and
relevant effects are determined to a large extent by the bioavailability of their gut-
derived metabolites. The human microbiota, composed of bacteria, fungi, archaea,
viruses, and protozoans, can be found throughout the body; as it colonizes the skin,
mouth, vagina, gastroenteric tube, and/or respiratory system. Over 70% of microbi-
ota colonizes the gastrointestinal tract and contains more than 100 trillion microbes.
The human microbiome, a synergistic community of microorganisms often con-
sidered an independent endocrine organ in a symbiotic relationship with the host, is
comprised of two primary phyla, namely Bacteroidetes and Firmicutes. These two
phyla represent 98% of the gut microbiota and are typically found in a ratio favor-
ing Bacteroidetes over Firmicutes (B/F >1) when we consider a healthy/desirable
gut phenotype. Notably, however, a universal definition of “healthy microbiota” as
it would pertain to a specific microbial profile is still lacking. Other phyla include
Proteobacteria, Verrucomicrobia, Fusobacteria, Cyanobacteria, and Actinobacteria.
Firmicutes can be grouped into three major classes: Clostridia, Negativicutes, and
Bacilli, while Firmicutes overall consist of over 200 genera, including Staphylococcus,
Lactobacillus, Ruminococcus, and Clostridium. The relevant phyla consist primar-
ily of gram-positive bacteria, except for those belonging to the Negativicutes class.
Powerful Interplay of Gut Microbiota and Polyphenols 191
Monomers/Dimers Polymers
Phase II
xenobiotic
metabolism
FIGURE 7.2 Conceptual schematic depicting the major bio-transformations and relevant
locations of dietary polyphenols (the blue dashed line represents separation between the intes-
tinal lumen and the internal organismal space).
microbial metabolites could be among the new generation of therapeutic agents for
the management of immunoinflammatory diseases such as atherosclerosis, but also
for stifling the inflammatory response to bacterial antigens, which plausibly extends
significant implications in terms of chronic inflammatory or autoimmune diseases
such as inflammatory bowel disease (Cardona et al., 2013).
Interestingly, Beloborodova et al. analyzed the role of phenolic acids of microbial
origin as biomarkers in the progress of sepsis. They reported that p-hydroxyphenylacetic
acid was able to prevent reactive oxygen species production in neutrophils. By acting on
neutrophils, there is retardation of immune responses, however, when acting on mito-
chondria, there is preclusion or attenuation of multiple organ failure. Thus, during the
development of bacteremia and purulent foci of infection associated with P. aerugi-
nosa and Acinetobacter baumanii, their metabolite, p-hydroxyphenylacetic acid, can
directly enter the systemic blood flow and inhibit the phagocytic activity of neutrophils
(Beloborodova et al., 2012).
While limited studies are investigating these relationships in humans, some rep-
resentative characteristic examples are discussed below. In healthy men, a single
intake of approximately 240 g of fresh blueberries, rich in polyphenols such as
anthocyanins, was shown to increase flow-mediated dilation (FMD), accompanied
by increases in plasma concentrations of phenolic metabolites such as vanillic acid,
homovanillic acid, benzoic acid, hippuric acid, and hydroxyhippuric acid. Hence,
sufficient residence time of blueberry metabolites in the circulation allows for their
vascular activity to be exerted. In addition, blueberries induced a dose-dependent
and biphasic increase in FMD, while the elevation in plasma polyphenol metabolites
occurred in tandem with FMD improvement (Rodriguez-Mateos et al., 2013).
Dietary intervention with 66 healthy men consuming polyphenols from Aronia
berry (extract or fruit) for 12 weeks demonstrated significant associations between
changes in endothelial function, plasma metabolites of Aronia berry polyphe-
nols, and specific gut microbial genera. FMD, used to assess endothelial function,
increased alongside changes in the gut microbiota. Aronia (poly)phenol-rich extract
(116 mg, 75 g berries) was shown to increase the growth of Anaerostipes and whole
fruit powder (12 mg, 10 g berries) caused a notable increase in Bacteroides, without
any changes in the diversity of gut microbiota in either treatment (Istas et al., 2019).
The authors concluded that in healthy men, Aronia consumption resulted in improved
endothelial function and positively modulated gut microbiota composition, indicat-
ing a potential benefit for maintaining cardiovascular health (Istas et al., 2019).
Moreover, A. muciniphila was reported to be induced post-consumption of
pomegranate extract (rich in hydrolyzable ellagitannins) in 16 out of the 20 healthy
participants that were producing urolithin-A (Li et al., 2015). Nevertheless, in a
separate study with 49 healthy overweight or obese participants consuming a higher
dose of a different pomegranate extract, no significant alterations were observed for
A. muciniphila (Gonzalez-Sarrias et al., 2017). A clinical trial with obese insulin-
resistant patients showed that resveratrol, a natural phenolic compound, increased
the abundance of A. muciniphila in Caucasians but not in other ethnic groups
(Verhoog et al., 2019). In conclusion, there is a variety of gut bacteria that can pro-
duce metabolic by-product compounds when exposed to polyphenols and may pro-
mote health in humans.
Powerful Interplay of Gut Microbiota and Polyphenols 197
There is a series of recent human studies that aimed to use different approaches to
delineate the effects of plant polyphenols on the gut microbiota. Basak et al. recently
studied the efficacy of curcumin on tumor suppression. More specifically, a double-
blind, randomized, placebo-controlled phase 1 clinical trial was conducted with
APG-157 in 13 normal subjects and 12 patients with oral cancer. Two doses of 100 or
200 mg were delivered transorally every hour for 3 hours. Blood and saliva were col-
lected before and 1, 2, 3, and 24 hours after treatment. Electrocardiograms and blood
tests did not demonstrate any toxicity. Curcumin was found in the blood and tumor
tissues. Inflammatory markers and Bacteroides species were found to be decreased
in the saliva, and immune T-cells were increased in the tumor tissue. APG-157 is
absorbed well, reduces inflammation, and attracts T-cells to the tumor, suggesting
its potential use in combination with immunotherapy drugs. Though endowed with
properties of tumor cell suppression due to its antioxidant and anti-inflammatory
actions, curcumin is poorly absorbed when administered orally and consequently
less effective. APG-157, a botanical drug containing curcumin among other poly-
phenols, on the other hand, is absorbed well and is reported to be potentially benefi-
cial when combined with immunotherapy by reducing inflammation and increasing
T-cell concentration in the tumor (Basak et al., 2020).
Vetrani and colleagues showed that a diet naturally rich in polyphenols and/or
long-chain n − 3 polyunsaturated fatty acids (LCn3) increased the diversity of the
predominant fecal bacteria; the polyphenols increased Clostridium leptum (clostrid-
ial cluster IV), which was directly associated with good glucose tolerance, by early
secretion of insulin. More specifically, 78 individuals with high waist circumference
and at least one additional component of the metabolic syndrome were randomized
to an isoenergetic 8-week diet: (a) low LCn3 and polyphenols; (b) high LCn3; (c) high
polyphenols; or (d) high LCn3 and polyphenols. Microbiota analysis was performed
on feces collected before and after the intervention. Denaturing gradient gel elec-
trophoresis (DGGE) analysis of the predominant bacteria, Eubacterium rectale and
Blautia coccoides group (Lachnospiraceae, EREC), C. leptum (Ruminococcaceae,
CLEPT), Bacteroides spp., Bifidobacteria, and Lactobacillus group was performed.
A quantitative real-time PCR was performed for the same group additionally includ-
ing the Atopobium cluster (Coriobacteriaceae). Before and after the intervention,
participants underwent a 75 g oral glucose tolerance test and a high-fat test meal
to evaluate glucose and lipid response. The authors reported that polyphenols sig-
nificantly increased microbial diversity and CLEPT but reduced EREC. LCn3 sig-
nificantly increased the numbers of Bifidobacteria. Τhus, it was concluded that diets
naturally rich in polyphenols or LCn3 influenced gut microbiota composition in
individuals at high cardiometabolic risk, with these modifications associated with
changes in glucose/lipid metabolism (Vetrani et al., 2020).
In an in vitro colon system study, polyphenol-rich fractions of blueberry-con-
taining anthocyanins/flavonol glycosides (ANTH/FLAV), PACs, sugar/acid frac-
tion (S/A), and total polyphenols (TPP) had a distinct effect on fecal microbiota
composition. Effective promotion of microbiome alpha diversity was observed with
ANTH/FLAV, and PAC fractions as opposed to the S/A and TPP fractions, which
has been attributed to the differentially responsive taxa. Older compared to the
younger group showed an abundance of gut microbiota diversity with blueberrry
198 The Gut Microbiome: Bench to Table
microbiota found that the composition of total bacteria was similar. A reduction
in ammonia and an increase in the production of SCFAs were also elucidated. The
study implies a positive effect on the daily consumption of orange juice in young
women (Lima et al., 2019).
Consumption of an olive pomace-enriched biscuit formulation (OEP), which
delivers 17.1 ± 4.01 mg/100 g of hydroxytyrosol and its derivative on analysis, showed
an upregulation of the microbial polyphenol biotransformation in the intestine, as
evidenced by a significant increase in the excretion of small phenolic acids in urine.
OEP also significantly elevated homovanillic acid and 3,4-dihydroxyphenylacetic
acid in fasted plasma samples, indicating clearance of these compounds from blood
and an extended release and uptake from the intestine. However, the study failed to
show any change in ox-LDL or urinary isoprostane (Conterno et al., 2019).
A study on the targets of curcumin natural polyphenols on nonalcoholic fatty liver
disease determined amino acids, TCA cycle, bile acids, and gut microbiota. Phospholipid
curcumin supplement led to a significant reduction in 3-methyl-2-oxovaleric acid,
3-hydroxyisobutyrate, kynurenine, succinate, citrate, α-ketoglutarate, methylamine, tri-
methylamine, hippurate, indoxyl sulfate, chenodeoxycholic acid, taurocholic acid, and
lithocholic acid (Chashmniam et al., 2019).
Dietary intervention with functional foods in patients with T2DM who exhibit
intestinal dysbiosis, characterized by an increase in Prevotella copri, showed a
reduction in P. copri and increased species with anti-inflammatory effects, namely
Faecalibacterium prausnitzii and A. muciniphilia. A significant reduction in area
under curve for glucose, total and LDL cholesterol, FFA, HbA1c, triglyceride, and
CRP, as well as an increase in antioxidant activity was also observed. The study sug-
gested a beneficial effect on fecal microbiota, pointing to novel avenues for improving
glycemic control, dyslipidemia, and inflammation with long-term adherence to high-
fiber, polyphenol-enriched, vegetable-protein-based diet (Medina-Vera et al., 2019).
In another study, Kim and colleagues showed that mango (Mangifera indica L.)
polyphenols reduce IL-8, growth-related oncogene (GRO), and GM-SCF plasma
levels and increase Lactobacillus species in a pilot study in patients with inflam-
matory bowel disease (Kim et al., 2020). In this study, ten participants received
a daily dose of 200–400 g of mango pulp for 8 weeks. Mango intake significantly
improved the primary outcome, Simple Clinical Colitis Activity Index score, and
decreased the plasma levels of pro-inflammatory cytokines including interleukin-8
(IL-8), GRO , and granulocyte macrophage colony-stimulating factor by 16.2%
(p = 0.0475), 25.0% (p = 0.0375), and 28.6% (p = 0.0485), all factors related to neu-
trophil-induced inflammation, respectively. Mango intake beneficially altered fecal
microbial composition by significantly increasing the abundance of Lactobacillus
spp., Lactobacillus plantarum, Lactobacillus reuteri, and Lactobacillus lactis,
which was accompanied by increased fecal butyric acid production. Therefore, an
enriching diet with mango fruits or potentially other gallotannin-rich foods seems
to be a promising adjuvant therapy combined with conventional medications in the
management of IBD via reducing biomarkers of inflammation and modulating the
intestinal microbiota (Kim et al., 2020).
Yang and colleagues studied the effect of standardized grape powder consump-
tion, a rich source of polyphenols and fiber, on the gut microbiome of healthy par-
ticipants. More specifically, 46 g of whole grape powder, providing the equivalent
200 The Gut Microbiome: Bench to Table
of two servings of California table grapes, were assessed as per their effects on the
gut microbiome in healthy adults. The study included a 4-week standardization to a
low-polyphenol diet, followed by 4 weeks of 46 g of grape powder consumption while
continuing the low-polyphenol diet. Compared to the baseline, 4 weeks of grape pow-
der consumption significantly increased the alpha diversity index of the gut microbi-
ome. The authors concluded that grape powder consumption significantly modified
the gut microbiome (Yang et al., 2021).
In separate experiments through a small-scale pilot study, Ezzat-Zadeh and cowork-
ers reported that California strawberry consumption increased the abundance of gut
microorganisms related to lean body weight, health, and longevity in healthy partici-
pants (Ezzat-Zadeh et al., 2021). More specifically, 15 healthy adults consumed a beige
diet + 26 g of freeze-dried strawberry powder (SBP) for 4 weeks, followed by 2 weeks
of beige diet only. Stool samples were collected at 0, 4, and 6 weeks. Fecal microbiota
was analyzed by 16S rRNA sequencing. Fecal cholesterol, bile acid (BA), and micro-
bial metabolites were assessed via gas chromatography. Daily SBP altered the abun-
dance of 24 operational taxonomic units (OTUs). Comparing week 4 to baseline, the
most significant increases were observed for one OTU from Firmicutes\Clostridia\
Christensenellaceae\NA, one OTU from Firmicutes\Clostridia\Mogibacteriacea\NA,
one OTU from Verrucomicrobia\Verrucomicrobiaceae\Akkermansia muciniphila,
one OTU from Actinobacteria\Bifidobacteriaceae\Bifidobacterium\NA, and one OTU
from Bacteroidetes\Bacteroidia\Bacteroidaceae\Bacteroides and a decrease of one
OTU from Proteobacteria\Betaproteobacteria\Alcaligenaceae\Sutterella. Comparing
weeks 4–6, we observed a reversal of the same OTUs from C Christensenellaceae,
V muciniphilia, and C Mogibacteriaceae. Fecal SCFAs and most of the fecal mark-
ers including cholesterol, coprostanol, and primary and secondary BAs were not
changed significantly except for lithocholic acid, which was increased significantly
at week 6 compared to baseline (Ezzat-Zadeh et al., 2021).
Wilson and collaborators investigated SunGold kiwi fruit supplementation of
individuals with prediabetes and demonstrated that such supplementation alters gut
microbiota and improves vitamin C status as well as anthropometric and clinical
markers. More specifically, two SunGold kiwifruit per day over 12 weeks were con-
sumed by participants (n = 24). Venous blood samples were collected at each study
visit (baseline, 6, and 12 weeks) to determine glycemic indices, plasma vitamin C
concentrations, hormones, lipid profiles, and high-sensitivity CRP. Participants pro-
vided a fecal sample at each study visit. DNA was extracted from the fecal sam-
ples and a region of the 16S ribosomal RNA gene was amplified and sequenced to
determine fecal microbiota composition. Compared to a baseline measurement, the
results of 12 weeks showed a significant increase in plasma vitamin C (14 µmol/L,
p < 0.001). There was a significant reduction in both diastolic (4 mmHg, p = 0.029)
and systolic (6 mmHg, p = 0.003) blood pressure and a significant reduction in waist
circumference (3.1 cm, p = 0.001) and waist-to-hip ratio (0.01, p = 0.032). Results also
showed a decrease in HbA1c (1 mmol/mol, p = 0.005) and an increase in fasting
glucose (0.1 mmol/L, p = 0.046); however, these changes were small and were not
clinically significant (Wilson et al., 2018).
An interesting study investigating the effects of a dietary supplement on the
gut microbiota was a recent study by Shin and colleagues (Shin et al., 2021). The
Powerful Interplay of Gut Microbiota and Polyphenols 201
Even though there is a significantly large body of evidence in support of the gut
microbiome’s role and specific enzymes in the biotransformation of polyphenols,
there are still significant aspects of the interplay between the microbiome and poly-
phenol metabolism that need to be unlocked. It appears that gut bacteria possess
innate mechanisms, allowing them to generate de novo and potentially highly bioac-
tive compounds when exposed to dietary polyphenols.
In this regard, innovative approaches are essential to reveal the precise activities
of certain bacteria and the exact role they play in the biotransformation of poly-
phenols. One such approach entails gut bacterial population characterization, using
metagenomic binning of bacterial DNA methylation to create bacterial barcodes for
efficient identification of unique bacterial strains. Other efforts employ gnotobiotic
and germ-free mice to manipulate bacterial populations so as to better understand
host metabolism. Such novel approaches can enhance identification of the distinc-
tive metabolism of gut microbiota, different among individuals, as well as elucidate
how this may regulate bioavailability and modulate the bioactivity of polyphenolic
metabolites (Pasinetti et al., 2018). These approaches could provide more insights
as to the interpersonal variation usually observed in the microbiome and metabolite
types in human trials. Such complex polyphenol–microbiota interactions, which are
determined notably by interindividual variability leading to different polyphenol-
metabolizing phenotypes or “metabotypes”, may be the key in terms of a more
202 The Gut Microbiome: Bench to Table
personalized approach for the optimization of diet and health with the involvement
of the microbiome (Bolca et al., 2013). The term “metabotype” refers to a particu-
lar metabolic phenotype with specific gut microbiome-derived metabolites, which
characterize metabolism of the parent compound (i.e., nutrient, pharmaceutical, and
toxin) (Espin et al., 2017).
In general, limited human studies have studied metabotypes. More specifically, a
pilot study assessed the phenolic compound capsaicin and reported stronger effects
on metabolic and inflammatory markers in healthy participants of the Bacteroides
enterotype compared to those of the Prevotella enterotype (Kang et al., 2016). Other
researchers reported that an interaction was observed between gut bacterial profiles
and enterolignan production in response to lipid levels after 1 week of intervention
in healthy normal-weight participants (Lagkouvardos et al., 2015). Interestingly,
even though the ability to stratify the population based on their capacity to pro-
duce specific microbial-derived metabolites in response to certain polyphenols and
the impact of this differential on health has not been investigated significantly,
most available evidence pertains to the correlation of certain metabotypes to iso-
flavones and ellagitannins. More specifically, randomized clinical trials have dem-
onstrated that patients’ capacity to produce the microbial metabolites equol and
O-desmethylangolesin (ODMA) has been shown to reduce the risk for CVD (Espin
et al., 2017). Ellagitannins represent another interesting example of a nexus between
a specific metabotype and a polyphenolic compound(s). In this regard, there are
three distinct metabotypes: (a) type A which is characterized by excretion of Uro-A
and related conjugates as the final urolithin; (b) type B which produces Uro-B and/
or IsoUro-A additionally to Uro-A; and (c) type 0 which does not produce any of
these urolithins. Interestingly, metabotype B has been correlated with obesity, meta-
bolic syndrome, and colorectal cancer, hence suggesting a potential role in dysbiosis
for this metabotype (Espin et al., 2017). Furthermore, in certain human trials with
overweight participants, while metabotype A was positively associated with HDL
cholesterol, metabotype B was associated with VLDL (Selma et al., 2009). These
interesting results could at least partly contribute to a potential explanation for mixed
and nonconclusive results in the case of polyphenol-rich dietary supplementations
such as the earlier case of the pomegranate study, for example. Interindividual vari-
ability regarding the improvement of cardiovascular risk in 49 healthy overweight
or obese participants consuming pomegranate was explained by urolithin metabo-
types (Gonzalez-Sarrias et al., 2017). Although the exact mechanisms explaining the
gut microbiota/polyphenol interplay-related impact on human health remains under
investigation, it appears that microbiota-derived bioactive metabolites from dietary
polyphenols are primarily involved in such effects.
Finally, it is important to consider the overall diet in addition to the interventions
made in terms of polyphenols when microbiota is considered. There is strong evi-
dence supporting the plasticity of the microbiome and it is significantly influenced
by the dietary regimen of the individual regardless of other factors. For example, in a
cross-sectional study, Noh et al. investigated the association of habitual dietary intake
with the taxonomic composition and diversity of the human gut microbiota among
222 Koreans aged 18–58 years (Noh et al., 2021). Gut microbiota data were obtained
by 16S rRNA gene sequencing on DNA extracted from fecal samples. The habitual
Powerful Interplay of Gut Microbiota and Polyphenols 203
diet for the year previous to the study was evaluated through a food frequency ques-
tionnaire. After multivariable adjustment, intake of several food groups, including
vegetables, fermented legumes, legumes, dairy products, processed meat, and nonal-
coholic beverages, were associated with major phyla of the gut microbiota. A dietary
pattern related to higher α-diversity (HiαDP) derived by reduced rank regression
was characterized by higher intakes of fermented legumes, vegetables, seaweeds,
and nuts/seeds and lower intakes of nonalcoholic beverages. The HiαDP was found
to be positively associated with several genera of Firmicutes such as Lactobacillus,
Ruminococcus, and Eubacterium (p <0.05). Among enterotypes identified by prin-
cipal coordinate analysis based on the β-diversity, the Ruminococcus enterotype
exhibited higher HiαDP scores and was strongly positively associated with the intake
of vegetables, seaweeds, and nuts/seeds, compared to the two other enterotypes. The
authors concluded that a plant- and fermented food-based diet was positively associ-
ated with certain genera of Firmicutes indicative of improved gut microbial health.
Their conclusions indicate that diet is a very strong determinant, in its own right, of
the microbiota profile in humans, which should be appreciated and considered as
such in studies and therapeutic approaches.
Moreover, significant in size, human studies with long-term assessment of diet also
support further the point discussed above. More specifically, Yu et al. showed that
long-term diet quality is associated with gut microbiome diversity and composition in
urban Chinese adults (Yu et al., 2021). The studies comprised 1,920 men and women,
enrolled in two prospective cohorts (baseline 1996–2006), who remained free of
CVDs, diabetes, and cancer at stool collection (2015–2018) and had no diarrhea or
antibiotic use in the last 7 days before stool collection. The microbiome was profiled
by 16S rRNA sequencing. Long-term diet was assessed by repeated surveys at baseline
and follow-ups (1996–2011), with intervals of 5.2–20.5 years between dietary surveys
and stool collection. Associations of dietary variables with microbiome diversity and
composition were evaluated by linear or negative binomial hurdle models, adjusting
for potential confounders. False discovery rate (FDR) <0.1 was considered significant.
Diet quality was positively associated with microbiome α-diversity and abundance of
Firmicutes, Actinobacteria, Tenericutes, and genera/species within these phyla, includ-
ing Coprococcus, Faecalibacterium/Faecalibacterium prausnitzii, Bifidobacterium/
Bifidobacterium adolescentis, and order RF39 (all FDRs <0.1). Significant associations
were also observed for intakes of dairy, fish/seafood, nuts/legumes, refined grains, and
processed meat, including a positive association of dairy with Bifidobacterium and
inverse associations of processed meat with Roseburia/Roseburia faecis. Most asso-
ciations were similar, with or without adjustment for BMI and hypertension status or
excluding participants with antibiotic use in the past 6 months (Yu et al., 2021).
Overall, there are limited studies in humans, typically with a small number of
participants examining the effect of polyphenols on the configuration and activity of
the gut microbiome. This represents an important gap in our understanding regarding
the polyphenol–microbe interactions in humans. Nonetheless, there is some evidence
mostly in the form of in vitro, in vivo studies, while more rarely clinical studies,
suggesting that particular doses of specific polyphenols do extend modulation of the
gut microbial populations; hence, as certain bacterial groups can be inhibited, oth-
ers can thrive. While this concept carries biological plausibility, further research is
204 The Gut Microbiome: Bench to Table
warranted and more results need to be obtained for such a relationship to be estab-
lished and most critically a mechanistic explanation to be derived.
In conclusion, all the findings suggest that in addition to the dietary polyphenols,
their microbial metabolites must also be taken into consideration when assessing
the impact of polyphenols on the host’s health. Despite the recent surge of inter-
est in their relationship and our increased appreciation of this dynamic cross talk
between dietary polyphenols and gut microbiota, we are far from fully grasping their
therapeutic potential for employing informed strategies in medical nutrition therapy
and/or microbial therapeutic interventions. However, smartly manipulating the gut
microbiota through dietary interventions for the prevention or treatment of disorders
might lead to the enrichment of “personalized nutrition” to be more comprehensive
in its considerations and help better understand the effects of dietary bioactive com-
pounds on the host microbiome.
From a clinical perspective and through a lens of developing therapeutic pharma-
ceuticals and schemes, there is strong evidence from human studies that polyphenols
contribute to disease prevention and the overall properties (antioxidant, anti-inflam-
matory) contribute to the favorable health outcomes documented (Safe et al., 2021).
However, it is equally evident from mechanistic in vitro and in vivo studies that
individual compounds as well as their mixtures modulate activity and/or expression
of multiple genes and downstream responses at various levels and degrees. For exam-
ple, similarly to other flavonoids, genistein interacts directly with multiple tyrosine
kinases, G-protein coupled receptors, and intracellular receptors alike. Therefore,
a potential clinical application utilizing genistein that would target a single gene/
pathway is rather complicated due to its diverse activities and highly possible off-
target effects. Discerning the successful development of clinical applications of
polyphenols, a series of requirements pertinent to mechanistic precision medicine
approaches need to be considered and include the following aspects:
and subsequently the bioavailability that ultimately affect the phenolic compounds
and their metabolites. They can directly act on the epithelium to modulate local
immune system functions and absorb metabolites into the systemic circulation
to have systemic effects on the host. It is a very challenging task for the research
community to grasp the relationship fully and delineate the various interplays
among the complex gut microbiota, the diverse types, and forms of dietary poly-
phenols, the varied immunomodulatory ability of the hosts, and connections with
gut–brain functions.
Innovative approaches such as that of Foodomics, driven by the integration of
the use of advanced-omics technologies, such as transcriptomics, proteomics, and
metabolomics, together with biostatistics, chemometrics, and bioinformatics, to
allow the evaluation of complex biological systems, as well as the mechanisms
of bioactive food compounds that may affect them, are emerging (Valdés et al.,
2022). The utilization of such methods has brought a significant change in the field
of food science, as the performed research may be redirected to ascertain new-
fangled associations. For example, by way of Foodomics-related applications, our
knowledge regarding the binomial between food/diet and health has widened.
Foodomics expand the possibilities toward unraveling the enormous complexity
of the Foodome, which has been defined as the pool of all compounds present in
a food item and/or in a biological system interacting with the investigated food
at a given time (Valdés et al., 2022). The investigation in foods of bioactive com-
pounds which widely vary in chemical structure, including polyphenols, can be
meaningfully supported by Foodomics toward understanding function, but also
investigating the presence, bioavailability, and biological characteristics (such as
toxicity, antioxidant, antiproliferative, or anti-inflammatory properties) of these
interesting molecules, while considering different food matrices and processing
influences (Valdés et al., 2022). Such approaches may constitute an important and
most helpful tool toward advancing our current understanding of those interac-
tions and their effects.
REFERENCES
Ángel García-Merino J, Moreno-Pérez D, de Lucas B, Montalvo-Lominchar MG, Muñoz E,
Sánchez L, Naclerio F, Herrera-Rocha KM, Moreno-Jiménez MR, Rocha-Guzmán NE,
Larrosa M. Chronic flavanol-rich cocoa powder supplementation reduces body fat mass
in endurance athletes by modifying the follistatin/myostatin ratio and leptin levels.
Food Funct. 2020;11(4):3441–3450. doi: 10.1039/d0fo00246a.
Aura A. Microbial metabolism of dietary phenolic compounds in the colon. Phytochem Rev.
2008;7:407–429. doi: 10.1007/s11101-008-9095-3.
Baenas N, Nuñez-Gómez V, Navarro-González I, Sánchez-Martínez L, García-Alonso J, Periago
MJ, González-Barrio R. Raspberry dietary fibre: Chemical properties, functional evalu-
ation and prebiotic in vitro effect. LWT. 2020;134:110140. doi: 10.1016/j.lwt.2020.110140.
Basak SK, Bera A, Yoon AJ, Morselli M, Jeong C, Tosevska A, Dong TS, Eklund M, Russ E,
Nasser H, Lagishetty V, Guo R, Sajed D, Mudgal S, Mehta P, Avila L, Srivastava M,
Faull K, Jacobs J, Pellegrini M, Shin DS, Srivatsan ES, Wang MB. A randomized,
phase 1, placebo-controlled trial of APG-157 in oral cancer demonstrates systemic
absorption and an inhibitory effect on cytokines and tumor-associated microbes. Cancer.
2020;126(8):1668–1682. doi: 10.1002/cncr.32644.
206 The Gut Microbiome: Bench to Table
Di Meo F, Donato S, Di Pardo A, Maglione V, Filosa S, Crispi S. New therapeutic drugs from
bioactive natural molecules: The role of gut microbiota metabolism in neurodegenera-
tive diseases. Curr Drug Metab. 2018;19:478–489.
Dueñas M, Muñoz-González I, Cueva C, Jiménez-Girón A, Sánchez-Patán F, Santos-
Buelga C, Moreno-Arribas MV, Bartolomé B. A survey of modulation of gut micro-
biota by dietary polyphenols. Biomed Res Int. 2015;2015:850902. doi: 10.1155/
2015/850902.
Edmands WM, Ferrari P, Rothwell JA, Rinaldi S, Slimani N, Barupal DK, Biessy C, Jenab M,
Clavel-Chapelon F, Fagherazzi G, Boutron-Ruault MC, Katzke VA, Kühn T, Boeing H,
Trichopoulou A, Lagiou P, Trichopoulos D, Palli D, Grioni S, Tumino R, Vineis P,
Mattiello A, Romieu I, Scalbert A. Polyphenol metabolome in human urine and its asso-
ciation with intake of polyphenol-rich foods across European countries. Am J Clin Nutr.
2015;102(4):905–913. doi: 10.3945/ajcn.114.101881.
Espín JC, González-Sarrías A, Tomás-Barberán FA. The gut microbiota: A key factor in the
therapeutic effects of (poly)phenols. Biochem Pharmacol. 2017;139:82–93. doi: 10.1016/
j.bcp.2017.04.033.
Ezzat-Zadeh Z, Henning SM, Yang J, Woo SL, Lee RP, Huang J, Thames G, Gilbuena I,
Tseng CH, Heber D, Li Z. California strawberry consumption increased the abundance
of gut microorganisms related to lean body weight, health and longevity in healthy sub-
jects. Nutr Res. 2021;85:60–70. doi: 10.1016/j.nutres.2020.12.006.
Filipski KK, Pacanowski MA, Ramamoorthy A, Feero WG, Freedman AN. Dosing recommen-
dations for pharmacogenetic interactions related to drug metabolism. Pharmacogenet
Genomics. 2016 Jul;26(7):334–339. doi: 10.1097/FPC.0000000000000220.
Filosa S, Di Meo F, Crispi S. Polyphenols-gut microbiota interplay and brain neuromodula-
tion. Neural Regen Res. 2018;13(12):2055–2059. doi: 10.4103/1673-5374.241429.
Gil-Sánchez I, Cueva C, Sanz-Buenhombre M, Guadarrama A, Moreno-Arribas MV,
Bartolomé B. Dynamic gastrointestinal digestion of grape pomace extracts: Bioaccessible
phenolic metabolites and impact on human gut microbiota. J. Food Compos. Anal. 2018;
68:41–52. doi: 10.1016/j.jfca.2017.05.005.
González-Sarrías A, García-Villalba R, Romo-Vaquero M, et al. Clustering according to
urolithin metabotype explains the interindividual variability in the improvement of
cardiovascular risk biomarkers in overweight-obese individuals consuming pome-
granate: A randomized clinical trial. Mol Nutr Food Res. 2017;61(5). doi: 10.1002/
mnfr.201600830.
Guevara-Cruz M, Godinez-Salas ET, Sanchez-Tapia M, Torres-Villalobos G, Pichardo-
Ontiveros E, Guizar-Heredia R, Arteaga-Sanchez L, Gamba G, Mojica-Espinosa R,
Schcolnik-Cabrera A, Granados O, López-Barradas A, Vargas-Castillo A, Torre-
Villalvazo I, Noriega LG, Torres N, Tovar AR. Genistein stimulates insulin sensitivity
through gut microbiota reshaping and skeletal muscle AMPK activation in obese subjects.
BMJ Open Diabetes Res Care. 2020;8(1):e000948. doi: 10.1136/bmjdrc-2019-000948.
Guglielmetti S, Bernardi S, Del Bo’ C, Cherubini A, Porrini M, Gargari G, Hidalgo-Liberona N,
Gonzalez-Dominguez R, Peron G, Zamora-Ros R, Winterbone MS, Kirkup B, Kroon
PA, Andres-Lacueva C, Riso P. Effect of a polyphenol-rich dietary pattern on intes-
tinal permeability and gut and blood microbiomics in older subjects: Study protocol
of the MaPLE randomised controlled trial. BMC Geriatr. 2020;20(1):77. doi: 10.1186/
s12877-020-1472-9.
Hidalgo-Liberona N, González-Domínguez R, Vegas E, Riso P, Del Bo’ C, Bernardi S, Peron G,
Guglielmetti S, Gargari G, Kroon PA, Cherubini A, Andrés-Lacueva C. Increased intes-
tinal permeability in older subjects impacts the beneficial effects of dietary polyphenols
by modulating their bioavailability. J Agric Food Chem. 2020;68(44):12476–12484. doi:
10.1021/acs.jafc.0c04976.
208 The Gut Microbiome: Bench to Table
Tresserra-Rimbau A, Lamuela-Raventos RM, Moreno JJ. Polyphenols, food and pharma. Current
knowledge and directions for future research. Biochem Pharmacol. 2018;156:186–195.
doi: 10.1016/j.bcp.2018.07.050.
Tsuji H, Moriyama K, Nomoto K, Akaza H. Identification of an enzyme system for daidzein-
to-equol conversion in Slackia sp. strain natts. Appl Environ Microbiol. 2012;78(4):
1228–1236. doi: 10.1128/AEM.06779-11.
Valdés A, Álvarez-Rivera G, Socas-Rodríguez B, Herrero M, Ibáñez E, Cifuentes A.
Foodomics: Analytical opportunities and challenges. Anal Chem. 2022;94(1):366–381.
doi: 10.1021/acs.analchem.1c04678.
van Duynhoven J, Vaughan EE, Jacobs DM, Kemperman RA, van Velzen EJ, Gross G, Roger
LC, Possemiers S, Smilde AK, Doré J, Westerhuis JA, Van de Wiele T. Metabolic fate of
polyphenols in the human superorganism. Proc Natl Acad Sci U S A. 2011;108(Suppl 1):
4531–4538 doi: 10.1073/pnas.1000098107.
Vazhappilly CG, Amararathna M, Cyril AC, Linger R, Matar R, Merheb M, Ramadan WS,
Radhakrishnan R, Rupasinghe HPV. Current methodologies to refine bioavailabil-
ity, delivery, and therapeutic efficacy of plant flavonoids in cancer treatment. J Nutr
Biochem. 2021;94:108623. doi: 10.1016/j.jnutbio.2021.108623.
Vendrame S, Guglielmetti S, Riso P, Arioli S, Klimis-Zacas D, Porrini M. Six-week con-
sumption of a wild blueberry powder drink increases bifidobacteria in the human gut.
J Agric Food Chem. 2011;59(24):12815–12820.
Vendrame S, Klimis-Zacas D. Anti-inflammatory effect of anthocyanins via modulation of
nuclear factor-κB and mitogen-activated protein kinase signaling cascades. Nutr Rev.
2015;73(6):348–358. doi: 10.1093/nutrit/nuu066.
Verhoog S, Taneri PE, Roa Díaz ZM, et al. Dietary factors and modulation of bacteria strains
of Akkermansia muciniphila and Faecalibacterium prausnitzii: A systematic review.
Nutrients. 2019;11(7):1565. Published 2019 Jul 11. doi: 10.3390/nu11071565.
Vetrani C, Maukonen J, Bozzetto L, Della Pepa G, Vitale M, Costabile G, Riccardi G,
Rivellese AA, Saarela M, Annuzzi G. Diets naturally rich in polyphenols and/or long-
chain n-3 polyunsaturated fatty acids differently affect microbiota composition in high-
cardiometabolic-risk individuals. Acta Diabetol. 2020;57(7):853–860. doi: 10.1007/
s00592-020-01494-9.
Vilela MM, Salvador SL, Teixeira IGL, Del Arco MCG, De Rossi A. Efficacy of green tea
and its extract, epigallocatechin-3-gallate, in the reduction of cariogenic microbiota in
children: A randomized clinical trial. Arch Oral Biol. 2020;114:104727. doi: 10.1016/
j.archoralbio.2020.104727.
Williamson G. The role of polyphenols in modern nutrition. Nutr Bull. 2017;42(3):226–235.
doi:10.1111/nbu.12278.
Williamson G, Clifford MN. Role of the small intestine, colon and microbiota in determining
the metabolic fate of polyphenols. Biochem Pharmacol. 2017;139:24–39. doi: 10.1016/
j.bcp.2017.03.012.
Wilson R, Willis J, Gearry RB, Hughes A, Lawley B, Skidmore P, Frampton C, Fleming E,
Anderson A, Jones L, Tannock GW, Carr AC. SunGold kiwifruit supplementation of
individuals with prediabetes alters gut microbiota and improves vitamin c status, anthro-
pometric and clinical markers. Nutrients. 2018;10(7):895. doi: 10.3390/nu10070895.
Yang J, Kurnia P, Henning SM, Lee R, Huang J, Garcia MC, Surampudi V, Heber D, Li Z.
Effect of standardized grape powder consumption on the gut microbiome of healthy
subjects: A pilot study. Nutrients. 2021;13(11):3965. doi: 10.3390/nu13113965.
Yu D, Nguyen SM, Yang Y, Xu W, Cai H, Wu J, Cai Q, Long J, Zheng W, Shu XO. Long-term
diet quality is associated with gut microbiome diversity and composition among urban
Chinese adults. Am J Clin Nutr. 2021;113(3):684–694. doi: 10.1093/ajcn/nqaa350.
Zhang PY. Polyphenols in health and disease. Cell Biochem Biophys. 2015;73(3):649–664.
doi: 10.1007/s12013-015-0558-z.
8 Diet, Obesity, and
Gut Microbiome
Yu-Chieh Cheng
Academia Sinica
CONTENTS
Introduction ............................................................................................................ 211
Gut Microbiota Formation and Characteristic ....................................................... 214
Gut Microbial Composition, Dynamics, and Function ..................................... 214
Diversity and Composition of Gut Microorganisms Implicated in
Host Health Maintenance .................................................................................. 214
Energy Metabolism and Gut Microbiota ............................................................... 217
Gut Microbiota in Obesity ................................................................................ 217
Regulation on Host Metabolism........................................................................ 218
Modulating Feed Behavior and Satiety ............................................................. 220
Modulating Feed Behavior by Gut–Brain Axis ................................................ 222
Dietary Modulating Gut Microbiota Composition ................................................ 223
Dietary Styles in Relation to Gut Microbial Composition ................................ 223
Macronutrients ..................................................................................................224
Carbohydrates...............................................................................................224
Fat ...................................................................................................................... 225
Protein ............................................................................................................... 225
Micronutrients ................................................................................................... 226
Applications ........................................................................................................... 226
Clinical Therapeutics ........................................................................................ 227
Probiotics and Prebiotics Intervention .............................................................. 227
Conclusion ............................................................................................................. 230
References .............................................................................................................. 230
INTRODUCTION
Obesity and overweight have become serious public health issues worldwide. The
population of obese individuals has shown rapid growth over the past four decades.
From 1975 to 2016, obesity prevalence demonstrated nearly tripling growth. In 2016,
more than 1.9 billion adults (18 years and older) were defined as overweight, and
650 million adults were obese (World Health Organization, 2020). Overweight and
obesity issues also reflect human physiological conditions that represent abnormal
or excessive fat accumulation. To define overweight and obesity in adults, body
mass index (BMI) is a typical approach that calculates the ratio of a person’s weight
expressed in kilograms divided by the square of his/her height expressed in meters.
For the BMI scale, according to the World Health Organization definition, over-
weight in adults is a BMI greater than or equal to 25, whereas obesity in adults is
a BMI greater than or equal to 30. BMI provides a useful and standardized mea-
sure that only needs simple and physiological data to readily classify overweight and
obese populations in adults (World Health Organization, 2020).
While there are complex and multiple reasons for obesity, the fundamental cause
of obesity is an energy imbalance when energy intake exceeds energy expenditure.
Macronutrients from dietary intake are used as a metabolic fuel and for maintaining
physiological function, while any excess calories promote the accumulation of adi-
pose tissue, leading to obesity. The most common obesogenic dietary pattern is that
of a high-fat and high-sugar diet. High–energy–density foods provide few portions
with extremely high calories, a condition that, when combined with lower levels of
physical activity due to an increase in sedentary behavior, leads to obesity. Excessive
energy intake is a cause of obesity but not the single obesogenic factor (Chooi et al.,
2019). Alterations in dietary and physical activity patterns are often linked to envi-
ronmental and societal changes as well as a lack of policies to support health and
agriculture, change the mode of transportation, urban planning, food overprocessing,
and lack of education.
Overweight and obesity have a detrimental relationship with human health, which
may lead to metabolic disorders such as type 2 diabetes, dyslipidemia, and hyper-
tension. Other systemic complications, cardiovascular diseases, and musculoskeletal
disorders are also related to obesity and overweight. There is evidence that obesity
may increase the risk of liver, gallbladder, and pancreatic cancers, as well as hema-
tological cancers and aggressive prostate cancer (Calle and Kaaks, 2004). Previous
reports revealed that obese patients exhibit a higher risk for hospitalization and
mechanical ventilation support due to insufficient expiratory reserve volume, func-
tional capacity, and respiratory system compliance. Thus, these functional disorders
can lead to significant increases in morbidity and mortality from COVID-19 (Caussy
et al., 2020; Dietz and Santos-Burgoa, 2020; Popkin et al., 2020).
Currently, the mortality of obese and overweight individuals is higher than
underweight individuals in the world. The high prevalence of obesity develops seri-
ous social problems and may squander unnecessary medical resources in society.
Fortunately, obesity and overweight rates could be attenuated through a healthy diet
and lifestyle, as well as increasing physical activity. According to the World Health
Organization suggestions, limiting energy intake from sugars and fats; increasing
fruit, vegetable, legume, whole grain, and nuts consumption; and enhancing regu-
lar physical activity frequency are advised to individuals to prevent overweight and
obesity. With obesity becoming a worldwide epidemic, insights into how obesity
contributes to metabolism and human homeostasis, as well as new interventions, are
urgently needed. Nowadays, numerous research teams are dedicated to inquiry on
the mechanism to provide more efficient resolutions for obesity.
Diet, Obesity, and Gut Microbiome 213
The gut microbiota composition is unique and has large diversity, which plays a vital
role in host metabolism, immunomodulation, and biosynthesis. The gut microbiota
involves not only nutrient absorption but also metabolite production, including lipids,
amino acids, bile acids, and short-chain fatty acids (SCFAs). Moreover, resident bac-
teria inhibit pathogen invasion by maintaining intestinal epithelium integrity, com-
peting for nutrients and space with pathogens by pH modification and antimicrobial
peptide secretions (Coyte and Rakoff-Nahoum, 2019). Microorganisms are classified
into phyla, classes, orders, families, genera, and species by using the taxonomy. The
dominant phyla in the gut microbes are Firmicutes, Bacteroidetes, Actinobacteria,
Proteobacteria, Fusobacteria, and Verrucomicrobia. Firmicutes and Bacteroidetes
account for the vast majority of microorganisms which are approximately 97% of the
gut microbiota. Over 200 genera of Firmicutes are found in this phylum, including
Lactobacillus, Bacillus, Clostridium, Enterococcus, and Ruminicoccus. In addition,
the Clostridium genus constituted 95% of the Firmicutes phylum. Bacteroides are
the most abundant and variable genera which contain Bacteroides and Prevotella.
The predominant genera in Bacteroidetes are Bacteroides and Prevotella. For
Actinobacteria, the most abundant genus is Bifidobacterium (Arumugam et al., 2011;
Rinninella et al., 2019; Rosenbaum et al., 2015).
The first understanding of this interaction relied on germ-free (GF) mice, which exist
in a sterile environment completely without microorganisms. It is illustrated that
GF animals had a lower body weight and had poorer intestinal function than con-
ventionally raised mice. An enlarged liver and lower concentrations of amino acids
in the gut were observed as well. Scientists used GF mice to colonize microbiota
from conventionally raised mice, and the results showed a significant increase in
body weight, fat accumulation, and relative insulin resistance (Bäckhed et al., 2004).
Interestingly, although body weight was increased, the energy expenditure grew by
approximately 30%, whereas energy intake declined by 30% approximately after
GF mice received a microbial community. Many GF animal studies proved resi-
dent microbes are implicated in the energy balance equation (Bäckhed et al., 2007;
Rosenbaum et al., 2015). An additional study mentioned that GF animals are pro-
tected against diet-induced obesity such as Western-style, high-sugar, and high-fat
diets (HFDs) (Bäckhed et al., 2007). Supporting this notion, increased expression of
glucagon-like peptide-1 (GLP-1), which is an obesity-related peptide in the brain and
can reduce food intake, was observed (Uzbay, 2019).
The species variation of gut microbiota is dynamic. The colonization of microbes
in the GI tract begins postpartum. Infants are born to be uncolonized with the gut
microbiota and then develop an unstable microflora receiving from the environment
and the maternal microbial reservoir. Infants temporarily acquire microbes and form
microflora from the maternal skin and vagina and then persistently accumulate from
Diet, Obesity, and Gut Microbiome 215
the maternal gut, and it has been proven that the strains are more adapted than other
sources (Ferretti et al., 2018). Many factors, such as types of birth delivery, gesta-
tional age at birth, methods of milk feeding, and antibiotic medications, impact the
composition of the neonatal gut microbiota. These pioneering microbes utilize lac-
tose, galactose, and sucrose as their energy sources; after infants switch diet into solid
food, the gut microbiome alternates to the pathway of carbohydrate fermentation and
vitamin biosynthesis (Zmora et al., 2019). However, dysbiosis has been revealed to
be related to several diseases during the early stage, such as allergies, asthma, type 1
diabetes, Crohn’s disease, IBD, necrotizing enterocolitis, and inflammation (Ferretti
et al., 2018; Renz and Skevaki, 2021). Developing a healthy gut microbial community
in the early stage is crucial for later life. Conversely, studies have indicated that the
human gut microbiome can be altered and develop into maturity over the different
life stages. Scientists have observed a decline in microbiota diversity during aging
in elderly people. There is a trend whereby saccharolytic bacteria decrease while
proteolytic bacteria increase (Bischoff, 2016). Elderly individuals have higher levels
of Escherichia coli, Bacteroidetes, and Proteobacteria than adults and infants but a
lower ratio of Firmicutes to Bacteroidetes than younger adults (Mariat et al., 2009).
The dominant population has been replaced by subdominant species, which may be
due to an age-related decrease in immune function and aging-associated pathologies
(Vaiserman et al., 2017).
Additionally, more factors alter the composition of the gut microbiome. Circadian
rhythms modulate daily events that affect feeding, sleeping, hormone secretion,
metabolism, and temperature, which rely on the oscillation of light during the 24 h
cycle (Liang and FitzGerald, 2017; Mu et al., 2016). The functions of the intestine, gut
motility, and nutrient absorption are also affected by the circadian rhythm (Hussain
and Pan, 2009). The gut microbiota shows diurnal variations modulated by circadian
rhythms. Bacteroidetes, Firmicutes, and Proteobacteria are reported to be related to
oscillation. Bacteroidetes were discovered to be the most sensitive phylum that exhib-
ited circadian rhythmicity. During the daytime, Bacteroidetes are more copious than
nighttime with abundance oscillations which impacts the gut microbiome assembly
(Liang et al., 2015; Zmora et al., 2019). In addition, disruption of the host circadian
clock drives loss of microbiota rhythms and dysbiosis which facilitates glucose intol-
erance and obesity in humans and mice (Thaiss et al., 2014).
Previous studies have recently shown the association between microbial profiles
and numerous human diseases, such as cardiovascular disease, IBD, irritable bowel
syndrome, obesity, and type 2 diabetes. Furthermore, accumulating evidence sug-
gests that the gut microbiota is involved in maintaining brain health. It has been
reported that the gut microbiota impacts the central nervous system homeostasis
indirectly by modulating neurotransmitter and host immune function and blood–
brain barrier integrity. Thus, the alterations react to stress responsivity, anxiety-like
behaviors, sociability, and cognition (Luczynski et al., 2016). Increasing validation
for the essential role of microbial metabolism in maintaining the host immune system
has been found in recent research analyzing the composition and function of individ-
ual microbial species and complex microbial communities. The crosstalk between
the gut microbiome and host immune system is complex and has systemic impacts
on the whole body. In general, resident bacteria produce metabolites by undigested
216 The Gut Microbiome: Bench to Table
Gut microbiota composition has been linked to BMI class and diet. The ratio of
Bacteroidetes and Firmicutes phyla shows variation in obese adults, in which
Bacteroidetes decrease dramatically and enrich the proportions of Firmicutes as
well as Proteobacteria. A clinical trial supports this notion. The individuals were
separated into two groups and received an underfeeding or overfeeding diet. The
results revealed that the energy intake has a positive correlation with the relative
amount of Firmicutes but a negative correlation with the relative abundance of
Bacteroidetes. These studies validated the gut microbiota is responsive to host energy
balance (Jumpertz et al., 2011). Moreover, scientists have discovered that differences
218 The Gut Microbiome: Bench to Table
at the genus and species level are more relevant to obesity. For example, the relative
amount of Bifidobacterium decreased while Lactobacillus increased conversely in
overweight to obese individuals (Million et al., 2012). Erysipelotrichaceae, the pre-
dominant family, has four groups of closely related species-level phylotypes that were
revealed to have different responses during dietary changes and metabolic pheno-
types (Zhao, 2013). Previous studies indicated lower proportions of Bifidobacterium
vulgatus in obese humans than in lean individuals (Bervoets et al., 2013; Rinninella
et al., 2019). These variations are very sensitive to energy balance and can be recov-
ered along with weight loss based on a healthy diet.
Similarly, the gene richness and diversity of the gut microbiome declines signifi-
cantly in obese adults compared to lean individuals and is enhanced after dietary
weight loss (Rosenbaum et al., 2015; Torres-Fuentes et al., 2017). Low microbial rich-
ness also has a relationship with host metabolic parameters, including serum insulin,
homeostasis model assessment insulin resistance, triglyceride levels, and free fatty
acids in plasma (Le Chatelier et al., 2013). This means that the variation in the gut
microbiota in obese populations results in dysbiosis and affects host metabolism and
health, which may lead to detrimental comorbidities and complications such as dia-
betes and metabolic syndrome. The low diversity of the gut microbiome also has a
high correlation with trunk fat mass and comorbidities such as type 2 diabetes and
hypertension (Aron-Wisnewsky et al., 2019). Furthermore, the obese microbiome is
transmittable. GF mice inoculated with bacteria from obese mice’s cecum exhibited
significant weight gain compared with those received from a lean donor. A decline
in fecal energy excretion is observed in colonization with microbiota from an obese
donor, and it has been illustrated that the gut microbiome from obese individuals can
enhance energy harvest capacity from the diet (Turnbaugh et al., 2006).
The crosstalk between the gut microbiome and host is implicated in metabolism and
energy homeostasis. Scientists have found this phenomenon by using the GF mouse
model. GF mice lacking gut microbiota are protected against diet-induced obesity
with an HFD. GF mice fed a lard-based HFD showed increased energy expenditure,
fecal fat content, and energy excretion, which led to diet-induced obesity. It has also
been observed that mice with microbiota depletion have more browning of the ingui-
nal subcutaneous and perigonadal visceral adipose tissue. This metabolic improve-
ment can enhance the volume of brown adipose tissue, improve insulin sensitivity, and
decrease white fat and adipocyte size (Suárez-Zamorano et al., 2015). In short, these
results indicate the reason why GF mice have the resistance to diet-induced obesity.
Additionally, accumulating evidence suggests that gut microbiome composition
and host metabolism are highly correlated. The alteration of the Bacteroidetes and
Firmicutes ratio in the gut depends on the difference in the host metabolism sta-
tus and it is illustrated that related to carbohydrate degradation and fermentation
(Ley et al., 2006). Moreover, scientists monitored the relationship between rest-
ing energy expenditure, body composition, and the gut microbiome. The popula-
tion of Firmicutes has demonstrated a positive correlation with fat mass, whereas
Diet, Obesity, and Gut Microbiome 219
the opposite relationship with resting energy expenditure and the maximal oxygen
consumption (VO2max) has been observed (Kocełak et al., 2013). The gut microbi-
ome composition can be altered by probiotics and prebiotic intervention. By using
inulin-type fructo-oligosaccharides as a supplement, an increase in health-promoting
species such as Bifidobacterium, Lactobacillus, Roseburia, and Faecalibacterium
has been observed. This alteration has been proven to decrease postprandial plasma
glucose responses after a standardized meal, also enhancing plasma GLP-1 and pep-
tide YY (PYY) concentrations which may contribute to host satiation and glucose
homeostasis (Cani et al., 2009).
Accumulating evidence proves that there are specific relevant functions in each
phylum. In Firmicutes, some species transfer indigested fiber into butyrate and join
symbiosis with pathogens. The other predominant phylum, Bacteroidetes, has been
reported to degrade polysaccharides and ferment fiber, and its major energy sources
are protein and grain-rich diets. Actinobacteria, which account for approximately 3%
of the total gut bacteria, have been illustrated to work on host vitamin biosynthesis.
In addition, although other minor phyla account for less than 1%, they are also impli-
cated in host metabolisms, such as iron degradation, vitamin synthesis, and carbohy-
drate fermentation (Consortium, 2012; Turnbaugh et al., 2009). Specific bacteria may
inhibit circulating lipoprotein lipase inhibitors in the intestinal epithelia, resulting in
host weight gain due to enhanced triglyceride synthesis, promoted fat accumulation
as well as reduced insulin sensitivity (Sun et al., 2018).
Furthermore, metabolites produced by resident microorganisms comprehensively
affect host energy homeostasis function. SCFAs, bacterial metabolites, provide an
important energy source for intestinal epithelial cells. The major consortiums of
bacteria that produce SCFAs were Faecalibacterium prausnitzii and Eubacterium,
Lactobacillus, and Bifidobacterium species. Bacteria produce SCFAs throughout
carbohydrate fermentation, and increased energy harvesting accounts for a sub-
stantial part of energy uptake. The gut microbiome also protects against Western
diet–induced obesity and metabolic disorders by SCFAs production. According
to previous studies, the intervention with butyrate and succinate in mouse models
improved glucose sensitivity and glucose tolerance and enhanced energy expenditure
(De Vadder et al., 2016; Gao et al., 2009). Type 2 diabetes patients have been discov-
ered with the α lower relative ratio of SCFA-producing bacteria in the gut microbiota.
Clinically, a high-fiber diet alters gut microbiota composition and improves patients’
glycated hemoglobin A1c (HbA1c) via induced GLP-1 production (Zhao et al., 2018).
Conversely, some studies have indicated that there is a negative correlation between
host metabolism and SCFAs, especially acetate and propionate. In the rodent model,
circulating acetate which is enriched by altering gut microbiota leads to activation of
the parasympathetic nervous system, which may drive insulin secretion and increase
the risk of obesity (Perry et al., 2016). Studies in humans have revealed that a diet
with propionate may induce insulin resistance and obesity. One of the reasons may
be that SCFAs are ligands of free fatty acid receptors (FFAR2 and FFAR3). Free
fatty acid receptors activation has a high correlation with satiety and insulin sensitiv-
ity (Tirosh et al., 2019). Hence, the beneficial effect of SCFAs on host metabolism is
controversial and inconsistent.
220 The Gut Microbiome: Bench to Table
et al., 2015). In addition, Heiss et al. indicated that gut microbiota enhances leptin
sensitivity by regulating GLP-1–dependent mechanism (Heiss et al., 2021).
Furthermore, ghrelin is a gastrointestinal peptide hormone secreted from the
stomach and stimulates appetite by affecting the hypothalamic arcuate nucleus
(Kojima and Kangawa, 2005). Serum ghrelin levels were proven to have a negative
correlation with the ratio of Bacteroidetes and Firmicutes, as well as the number of
Faecalibacterium and Prevotellaceae, and a positive correlation with the number of
total bacteria, Clostridium, and Ruminococcus. For Bacteroides, Bifidobacterium,
and Lactobacillus, both positive and negative correlations with ghrelin were observed
(Leeuwendaal et al., 2021; Queipo-Ortuño et al., 2013). Numerous studies have indi-
cated that microbial metabolites play a key role in ghrelin secretion. SCFAs are able
to suppress ghrelin secretion and reduce ghrelin receptor (the growth hormone secre-
tagogue receptor type 1a) stimulation; LPS has been proven to decrease circulating
ghrelin levels. On the contrary, amino acids have the capacity to promote ghrelin
release (Leeuwendaal et al., 2021).
The gut microbiome modulates the gut–brain function and occupies an important
modulator of the microbiota–gut–brain axis, which affects host energy homeostasis.
By using the GF animal model, scientists showed the first evidence of the absence
of microbiome implicated in brain regulation and altered animal behavior. Scientists
have gained more understanding of the neuron alterations by specific strains on
myelination, neurotransmitters, microglia, neurogenesis, dendritic growth, and the
blood–brain barrier. Numerous studies have found that the administration of probi-
otics and prebiotics can reduce negative impacts and deterioration of brain function
(Cryan et al., 2019). Moreover, recent research has indicated that the gut microbiome
highly participates in with satiety, digestive function, and further cognitive and psy-
chological effects. The gut conveys nutritional status signals with the central nervous
system and engages energy modulation by using enteroendocrine cells (EECs), the
vagus nerve, and the enteric nervous system (van Son et al., 2021). In addition, the
metabolites produced by microbes can be involved in regulating signal transduction.
Interplay between the gut and brain is crucial for host energy homeostasis. In
recent years, it has become clear that signals from the gut play a major role in
appetite regulation, energy balance, glucose homeostasis, and satiety. One of the
regulations might be the activation of neuroendocrine mechanisms. EECs, which
are located in the GI tract from the stomach to rectum, secrete gut hormones and
neurotransmitters such as somatostatin, serotonin, ghrelin, CCK, PYY, and GLP-1
that are stimulated by luminal nutrients and provide a wide range of human physi-
ological modulations. After secretion, these gut hormones are transported to the
target neuron or enteric nervous system and exert their effectiveness throughout
the bloodstream. CCK, GLP-1, PYY, and oxyntomodulin are known as modula-
tors of on postprandial satiety. During the fasting stage, the group of gut hormones
that were highly increased was featured in promoting food intake and inhibiting
gastric acid secretion, which included ghrelin, motilin, insulin-like peptide 5, and
somatostatin. Conversely, hormones which secret postprandial such as gastrin,
Diet, Obesity, and Gut Microbiome 223
histamine, GLP-1, and CCK are characterized by enhancing digest efficiency, pro-
moting satiety, and preparing for storage of nutrients (Gribble and Reimann, 2016;
Symonds et al., 2015; van Son et al., 2021).
Beyond gut hormones, neuronal orchestration has been revealed as a key element
in the gut–brain function. The vagus nerve is a mixed nerve that interfaces with the
parasympathetic control of the heart, lungs, and digestive tract. Appetite, gastroin-
testinal motility, and anti-inflammatory modulation are all associated with the vagus
nerve. In addition, gut hormones (ghrelin, PYY, CCK) can trigger the vagus nerve
directly. According to previous studies, obesity and an HFD reduce the sensitivity of
vagus nerve afferent fibers that respond to gut hormones. The gut microbiome influ-
ences the vagus nerve indirectly by driving EECs to stimulate neurotransmitters. In
addition, dysbiosis has proven to be correlated with vagus nerve innervation and sig-
naling by increasing LPS and inflammatory cytokines. (Sen et al., 2017) (Browning
et al., 2017; van Son et al., 2021). Moreover, microbial metabolites converted by
the gut microbiome, such as SCFAs, also play a crucial role in the gut–brain axis.
SCFAs are derivatives from undigested fiber that modulate satiety and may increase
energy expenditure. Additionally, a study revealed that SCFAs affect insulin signal-
ing which leads to inhibition of fat accumulation in adipose tissue and promotes lipid
and glucose metabolism in other tissues (Kimura et al., 2013). A portion of SCFAs
can interact with EECs and stimulate PYY and GLP-1 secretion (Larraufie et al.,
2018; Tolhurst et al., 2012; van Son et al., 2021).
Diet is a critical factor affecting the gut environment, gut transit time, pH, as well
as changes in the composition of gut microbiota and diversity that influence host
metabolism. An unhealthy style of diet may lead to microbiome dysbiosis and induce
several sequelae, including obesity and type 2 diabetes. (Kim et al., 2019). Thus, a
healthy diet could improve the composition of gut microbiota and reduce the risks of
host metabolic disorders. Previous studies have revealed the differences in diet pat-
terns such as Western diet, Mediterranean diet, vegetarian diet, and gluten-free diet
have their own gut microbiome profiles that have different impacts on host metabo-
lism (Lazar et al., 2019).
The Western diet, developed from the industrial evolution, is comprised of satu-
rated fats, sugar, salt, protein, additional additives, and lower fiber and micronutrients.
The Western diet has been proven to have a positive correlation with inflammation
that leads to metabolic disorders and obesity. The major gut microbiota profile of the
Western diet is the reduction in total bacterial volume and beneficial species includ-
ing Lactobacillus spp., Bifidobacterium spp., and Eubacterium spp. Due to suffi-
cient dietary fiber, some of the specific bacteria lack the sources to generate SCFAs.
The observation of SCFAs in the lumen is significantly lower when consuming a
Western diet that may enhance risks of inflammatory (Kim et al., 2019; Lazar et al.,
2019; Statovci et al., 2017). Conversely, vegan and vegetarian diets contain abundant
dietary fiber which is opposite to the Western diet. Dietary fiber provides a good
224 The Gut Microbiome: Bench to Table
energy source to beneficial bacteria that promote a stable gut microbiota profile and
increase the number of lactic acid bacteria. A low quantity of Bacteroides spp. and
Bifidobacterium spp. was observed in the group that consumed vegan or vegetarian
diets. In addition, these diets usually consume rich polyphenols which is from coffee,
artichokes, olive, asparagus, and berries, which can facilitate the growth of protec-
tive bacteria. Bifidobacterium and Lactobacillus which are known intestinal barrier
protectors, and butyrate-producing bacteria, including Faecalibacterium prausnitzii
and Roseburia, Bacteroides vulgatus, and Akkermansia muciniphila, are reported
to have an increase in vegan and vegetarian diets. Additionally, a decline in LPS
producers such as Escherichia coli and Enterobacter cloacae has also been observed
in vegan and vegetarian diets that lead to alleviation of inflammation (Glick-Bauer
and Yeh, 2014).
The gut microbiota profile of a gluten-free diet has a lower abundance of
Lactobacillus spp., Bifidobacterium spp., Ruminococcus bromii, and Roseburia faecis,
whereas the Victivallaceae and Clostridiaceae, E. coli, and total Enterobacteriaceae
demonstrate the opposite trend due to decreasing of polysaccharide and fiber intake
(De Palma et al., 2009). The gluten-free diet, which is characterized by a reduc-
tion or exclusion of alimentary fiber from specific foods (i.e., grain), now has been
considered as a therapeutic method for celiac disease (Reddel et al., 2019). The
Mediterranean diet consists of abundant vegetables, olive oil, legumes, fish, and a
low amount of red meat and dairy products that contain monounsaturated and poly-
unsaturated fatty acids as well as antioxidants and fiber. The gut microbiota profile
in the Mediterranean diet has a high amount of Lactobacillus spp., Bifidobacterium
spp., and Prevotella spp., but a low amount of Clostridium spp. This profile of the
gut microbiota can prevent obesity and improve lipid and cholesterol levels (Garcia-
Mantrana et al., 2018). Additionally, numerous studies have demonstrated that dif-
ference in the ratio intake of macronutrients and micronutrients can change the
composition of gut microbiota and diversity which can influence host metabolism.
Carbohydrates
Many research studies have highlighted the altered ability of carbohydrates. In gen-
eral, carbohydrates are separated into two types, which are digestible and nondigest-
ible carbohydrates, and the gut microbiota composition has different responses to
different types of carbohydrates. Starch and sugars such as glucose, lactose, fructose,
and sucrose constitute digestible carbohydrates. They release glucose into the circula-
tion and induce insulin to further respond after digestion in the intestine. The level of
glucose is positively correlated with Bifidobacteria and Prevotella but negatively cor-
related with Bacteroides (Holmes et al., 2012; Lazar et al., 2019). In addition, lactose
is related to the reduction of Clostridia (Francavilla et al., 2012). Furthermore, unlike
digestible carbohydrates, nondigestible carbohydrates are the main materials that
offer commensal bacteria to produce SCFAs. Quantity and types of fiber intake have
a huge effect on gut microbiota alternations. Negative association with Bacteroides
and Actinobacteria and positive association with Proteobacteria and Firmicutes were
discerned in fiber intervention, and Firmicutes and Actinobacteria were the main
Diet, Obesity, and Gut Microbiome 225
Fats are generally divided into several types based on their structure. Consumption
of saturated and trans fats has been proven to enhance the risks of cardiovascular
disease. Conversely, monounsaturated and polyunsaturated fats are known as health-
promoting fats that alleviate the risk of chronic diseases. In general, a positive corre-
lation with Bacteroides and Actinobacteria and a negative correlation with Firmicutes
and Proteobacteria were observed in the diet with rich fats. Fats from dairy prod-
ucts have been reported to promote delta Proteobacteria and induce inflammation
(Holmes et al., 2012). Dietary with rich fat modifies the gut microbiota structure with
abundant LPS-expressing bacteria that lead to enhanced circulating LPS levels in
humans (Amar et al., 2008). LPS is related to weight gain, adiposity accumulation,
and the induction of inflammatory markers in white adipose tissue that facilitate
host physiology to a preinflammatory status. It is interesting that the adverse effect
of saturated fat seems to be specific to mice fed a lard-rich diet, and meanwhile,
enrichment of Bacteroides, Turicibacter, and Bilophila spp. were observed to be
associated with the white adipose tissue inflammation, adiposity, and insulin intoler-
ance. In contrast, mice fed a diet with unsaturated fat exhibited increased amounts
of Bifidobacterium, Akkermansia, and Lactobacillus spp. with no metabolic disor-
ders observed (Caesar et al., 2015). Nevertheless, there are inconsistent results in the
human clinical trials and individuals who switch diet patterns from saturated fat to
unsaturated fat as caloric sources did not influence the gut microbiota structure but
reduced the total bacterial volume (Fava et al., 2013).
Protein
Contrarily, consumption protein presents a comprehensively different gut micro-
biota profile. Sources of protein are important characteristics to impact gut micro-
biota. Both the diet with vegetarian protein (whey and pea) and animal protein are
226 The Gut Microbiome: Bench to Table
Trace minerals and vitamins are known as micronutrients, and their intake is
crucial for physiological maintenance. Various studies have demonstrated the
interaction between micronutrients and gut microbiota and led to further meta-
bolic regulation. Iron supplementation suppresses the levels of Bifidobacterium
but enhances the counts of Bacteroides and E. coli. Another study revealed an
increase in the fecal Enterobacteriaceae family and a reduction in Lactobacillus.
These results are totally consistent with the argument that bacteria and some
pathogens are efficient iron scavengers. Thus, iron supplementation may give rise
to microbiota dysbiosis and pathogen invasion (Yilmaz and Li, 2018). Manganese
supplementation in mice increased the colonization of the heart and lethality of
Staphylococcus aureus infection, likely because the bacterium utilizes manganese
to protect itself from reactive oxygen species and neutrophil killing (Juttukonda
et al., 2017). Bacteroides vulgatus is a prominent responder to vitamin A, and
it increases in number in the absence of vitamin A. Bacteroides dorei decreases
when vitamin A is deficient (Hibberd et al., 2017). Vitamin D plays a vital role in
immune homeostasis maintenance due to its interaction with the gut microbiota.
It has been reported that vitamin D has a negative correlation with gram-negative
genera, such as Haemophilus and Veillonella. This variation may be promoted by
low intake or levels of vitamin D (Luthold et al., 2017).
APPLICATIONS
Cumulative evidence elucidates that gut microbiota composition and diversity influ-
ence host metabolism and has close relationships with obesity and metabolic disor-
ders. Currently, scientists gain more knowledge on the mechanism between resident
bacteria and host metabolism, which leads to the development of different ways to
manipulate gut microbiota, such as fecal microbiota transplantation, probiotics, and
prebiotic intervention. In addition, lifestyle and dietary patterns are well understood
to alter and optimize the gut microbiota profile, especially by enriching commensal
bacteria. In this section, we will review studies with different applications to gain
more comprehension on enhancing human wellness by fostering a healthy gut micro-
biota and environment.
Diet, Obesity, and Gut Microbiome 227
foods contain stable microorganisms, usually including diverse lactic acid bacteria
and their metabolites. The well-known fermented foods are yogurt, cheese, kefir,
cocoa, pickles, kimchi, sauerkraut, and red wine. The variety of bacteria that fer-
mented food contains may have health-promoting properties and modulated func-
tions on the gut microbiota. Probiotics in fermented food can interact with the host gut
microbiota and mediate the composition into a “healthy gut microbiota.” It has been
reported that dysbiosis can be prevented after consuming fermented food periodi-
cally (Stiemsma et al., 2020). Yogurt, fermented by cultured probiotics, ameliorates
human metabolic disorders, including insulin intolerance and hepatic fat fraction.
Additionally, a reduction in serum LPS levels and modulation of gut microbiota com-
position were observed in obese adults with metabolic syndrome and nonfatty liver
disease (Chen et al., 2019). The investigations of kefir were discrepancies about the
metabolism modulation. However, some studies have mentioned the improvement
of lipid metabolic parameters, fasting blood glucose, and HbA1c levels; in contrast,
body weight was not significantly different (Bourrie et al., 2020). Han et al. found
an increase in Bifidobacterium after kimchi consumption for 8 weeks, and the gene
correlated with metabolism, immunity, and digestion was upregulated in obese indi-
viduals (Han et al., 2015). For red wine, previous studies have revealed that the intake
of red wine increases Bifidobacterium, Prevotella, Bacteroides, Eggerthellalenta,
Enterococcus, Bacteroidesuniformis, and Blautiacoccoides/Eubacterium rectale
abundance in the gut microbiota. Meanwhile, a reduction in BMI and body weight
and amelioration of blood pressure, total cholesterol, high-density lipoprotein -cho-
lesterol levels are observed due to the rich polyphenols (Moreno-Indias et al., 2016;
Queipo-Ortuño et al., 2012).
Individuals with high-protein and low-carbohydrate diets have fewer lev-
els of Roseburia and Eubacterium rectale and lower levels of butyrate in their
feces, which may have detrimental effects (Russell et al., 2011). In addition, the
abundance of Prevotella and Bacteroides is increased after intake of red meat.
Furthermore, red meat contains L-carnitine, which induces the production of the
proatherogenic microbial metabolite trimethylamine N‐oxide production and may
induce the risk of metabolic syndrome (Lazar et al., 2019). HFDs have been posi-
tively correlated with metabolic syndrome and type 2 diabetes and have also been
known as obesogenic diets. In addition, saturated fats and trans fats are espe-
cially linked with cardiovascular disease. Consumption of monosaturated and
polyunsaturated fats can decrease the risk of chronic disease. For gut microbiota
modification, HFDs can enrich the numbers of Bacteroides and total anaerobic
microorganisms. In contrast, blooms of Bifidobacterium are observed after intake
of a low-fat diet and improve fasting glucose and total cholesterol (Fava et al.,
2013). As mentioned above, fruits and vegetables are full of dietary fiber, flavones,
and polyphenols. They are recommended for intake and bring multiple benefits
to optimize gut microbiota and GI tract health. A recent study used a multiomics
approach to demonstrate the intestinal microbiome response in vegetable-rich
diets. An increase in catalytic activities for carbohydrates and food proteins, an
increase of cell motility to access nutrients, and the release or synthesis of bio-
active metabolites and proteins that are beneficial to human health have been
observed (De Angelis et al., 2020).
230 The Gut Microbiome: Bench to Table
CONCLUSION
In this chapter, we elucidated the importance and characteristics of gut microbiota
and the interaction between bacteria and the host. In particular, we mentioned how
gut microbiota signals and contributes to host metabolism by microbial metabolites,
altering richness, and elevating or suppressing specific bacteria. The gut microbi-
ota plays a critical role in the host metabolism by modulating the gut environment,
immune system, metabolism, and appetite. The knowledge drives the development of
therapies that enhance human well-being by improving the gut microbiome composi-
tion and diversity. Thus, therapeutic applications of microbiota, including diet, probi-
otics, and prebiotics treatment, have become a novel approach to tackle the problem
of obesity and metabolic disorders.
REFERENCES
Abenavoli, L., Scarpellini, E., Colica, C., Boccuto, L., Salehi, B., Sharifi-Rad, J., Aiello, V.,
Romano, B., De Lorenzo, A., Izzo, A.A., Capasso, R., 2019. Gut microbiota and obesity:
a role for probiotics. Nutrients 11, 2690.
Alcock, J., Maley, C.C., Aktipis, C.A., 2014. Is eating behavior manipulated by the gastrointesti-
nal microbiota? Evolutionary pressures and potential mechanisms. Bioessays 36, 940–949.
Amar, J., Burcelin, R., Ruidavets, J.B., Cani, P.D., Fauvel, J., Alessi, M.C., Chamontin, B.,
Ferriéres, J., 2008. Energy intake is associated with endotoxemia in apparently healthy
men. The American Journal of Clinical Nutrition 87, 1219–1223.
Andreoli, M.F., Donato, J., Cakir, I., Perello, M., 2019. Leptin resensitisation: a reversion of
leptin-resistant states. Journal of Endocrinology 241, R81–R96.
Aron-Wisnewsky, J., Prifti, E., Belda, E., Ichou, F., Kayser, B.D., Dao, M.C., Verger, E.O.,
Hedjazi, L., Bouillot, J.-L., Chevallier, J.-M., 2019. Major microbiota dysbiosis in severe
obesity: fate after bariatric surgery. Gut 68, 70–82.
Arumugam, M., Raes, J., Pelletier, E., Le Paslier, D., Yamada, T., Mende, D.R., Fernandes,
G.R., Tap, J., Bruls, T., Batto, J.-M., 2011. Enterotypes of the human gut microbiome.
Nature 473, 174–180.
Bäckhed, F., Ding, H., Wang, T., Hooper, L.V., Koh, G.Y., Nagy, A., Semenkovich, C.F.,
Gordon, J.I., 2004. The gut microbiota as an environmental factor that regulates fat
storage. Proceedings of the National Academy of Sciences 101, 15718–15723.
Bäckhed, F., Manchester, J.K., Semenkovich, C.F., Gordon, J.I., 2007. Mechanisms under-
lying the resistance to diet-induced obesity in germ-free mice. Proceedings of the
National Academy of Sciences 104, 979–984.
Bernard, A., Ancel, D., Neyrinck, A.M., Dastugue, A., Bindels, L.B., Delzenne, N.M.,
Besnard, P., 2019. A preventive prebiotic supplementation improves the sweet taste per-
ception in diet-induced obese mice. Nutrients 11, 549.
Bervoets, L., Van Hoorenbeeck, K., Kortleven, I., Van Noten, C., Hens, N., Vael, C.,
Goossens, H., Desager, K.N., Vankerckhoven, V., 2013. Differences in gut micro-
biota composition between obese and lean children: a cross-sectional study. Gut
Pathogens 5, 1–10.
Bindels, L.B., Delzenne, N.M., Cani, P.D., Walter, J., 2015. Towards a more comprehensive
concept for prebiotics. Nature Reviews Gastroenterology & Hepatology 12, 303–310.
Bischoff, S.C., 2016. Microbiota and aging. Current Opinion in Clinical Nutrition &
Metabolic Care 19, 26–30.
Blaak, E., Canfora, E., Theis, S., Frost, G., Groen, A., Mithieux, G., Nauta, A., Scott, K.,
Stahl, B., van Harsselaar, J., 2020. Short chain fatty acids in human gut and metabolic
health. Beneficial Microbes 11, 411–455.
Diet, Obesity, and Gut Microbiome 231
Blaut, M., 2015. Gut microbiota and energy balance: role in obesity. Proceedings of the
Nutrition Society 74, 227–234.
Bourrie, B.C., Richard, C., Willing, B.P., 2020. Kefir in the prevention and treatment of obesity
and metabolic disorders. Current Nutrition Reports 9, 184–192.
Browning, K.N., Verheijden, S., Boeckxstaens, G.E., 2017. The vagus nerve in appetite regu-
lation, mood, and intestinal inflammation. Gastroenterology 152, 730–744.
Byrne, C., Chambers, E., Morrison, D., Frost, G., 2015. The role of short chain fatty acids in appe-
tite regulation and energy homeostasis. International Journal of Obesity 39, 1331–1338.
Caesar, R., Tremaroli, V., Kovatcheva-Datchary, P., Cani, P.D., Bäckhed, F., 2015. Crosstalk
between gut microbiota and dietary lipids aggravates WAT inflammation through TLR
signaling. Cell Metabolism 22, 658–668.
Calle, E.E., Kaaks, R., 2004. Overweight, obesity and cancer: epidemiological evidence and
proposed mechanisms. Nature Reviews Cancer 4, 579–591.
Campos, A.C., Rocha, N.P., Nicoli, J.R., Vieira, L.Q., Teixeira, M.M., Teixeira, A.L., 2016.
Absence of gut microbiota influences lipopolysaccharide-induced behavioral changes
in mice. Behavioural Brain Research 312, 186–194.
Cani, P.D., Bibiloni, R., Knauf, C., Waget, A., Neyrinck, A.M., Delzenne, N.M., Burcelin, R.,
2008. Changes in gut microbiota control metabolic endotoxemia-induced inflammation
in high-fat diet–induced obesity and diabetes in mice. Diabetes 57, 1470–1481.
Cani, P.D., Lecourt, E., Dewulf, E.M., Sohet, F.M., Pachikian, B.D., Naslain, D., De
Backer, F., Neyrinck, A.M., Delzenne, N.M., 2009. Gut microbiota fermentation of
prebiotics increases satietogenic and incretin gut peptide production with conse-
quences for appetite sensation and glucose response after a meal. The American
Journal of Clinical Nutrition 90, 1236–1243.
Caussy, C., Wallet, F., Laville, M., Disse, E., 2020. Obesity is associated with severe forms of
COVID-19. Obesity (Silver Spring, Md.), 28, 1175.
Chen, Y., Feng, R., Yang, X., Dai, J., Huang, M., Ji, X., Li, Y., Okekunle, A.P., Gao, G.,
Onwuka, J.U., 2019. Yogurt improves insulin resistance and liver fat in obese women
with nonalcoholic fatty liver disease and metabolic syndrome: a randomized controlled
trial. The American Journal of Clinical Nutrition 109, 1611–1619.
Cheng, F.-S., Pan, D., Chang, B., Jiang, M., Sang, L.-X., 2020. Probiotic mixture VSL# 3: an
overview of basic and clinical studies in chronic diseases. World Journal of Clinical
Cases 8, 1361.
Cheng, Y.-C., Liu, J.-R., 2020. Effect of Lactobacillus rhamnosus GG on energy metabolism,
leptin resistance, and gut microbiota in mice with diet-induced obesity. Nutrients 12, 2557.
Chooi, Y.C., Ding, C., Magkos, F., 2019. The epidemiology of obesity. Metabolism 92, 6–10.
Consortium, H.M.P., 2012. Structure, function and diversity of the healthy human microbi-
ome. Nature 486, 207.
Coyte, K.Z., Rakoff-Nahoum, S., 2019. Understanding competition and cooperation within
the mammalian gut microbiome. Current Biology 29, R538–R544.
Cryan, J.F., O‘Riordan, K.J., Cowan, C.S., Sandhu, K.V., Bastiaanssen, T.F., Boehme, M.,
Codagnone, M.G., Cussotto, S., Fulling, C., Golubeva, A.V., 2019. The microbiota-
gut-brain axis. Physiological Reviews. 99, 1877–2013.
David, L.A., Maurice, C.F., Carmody, R.N., Gootenberg, D.B., Button, J.E., Wolfe, B.E.,
Ling, A.V., Devlin, A.S., Varma, Y., Fischbach, M.A., 2014. Diet rapidly and reproduc-
ibly alters the human gut microbiome. Nature 505, 559–563.
Davis, L.M., Martínez, I., Walter, J., Goin, C., Hutkins, R.W., 2011. Barcoded pyrosequencing
reveals that consumption of galactooligosaccharides results in a highly specific bifido-
genic response in humans. PLoS One 6, e25200.
De Angelis, M., Ferrocino, I., Calabrese, F.M., De Filippis, F., Cavallo, N., Siragusa, S.,
Rampelli, S., Di Cagno, R., Rantsiou, K., Vannini, L., 2020. Diet influences the func-
tions of the human intestinal microbiome. Scientific Reports 10, 1–15.
232 The Gut Microbiome: Bench to Table
De Filippis, F., Pellegrini, N., Vannini, L., Jeffery, I.B., La Storia, A., Laghi, L., Serrazanetti, D.I.,
Di Cagno, R., Ferrocino, I., Lazzi, C., 2016. High-level adherence to a Mediterranean diet
beneficially impacts the gut microbiota and associated metabolome. Gut 65, 1812–1821.
De Palma, G., Nadal, I., Collado, M.C., Sanz, Y., 2009. Effects of a gluten-free diet on gut
microbiota and immune function in healthy adult human subjects. British Journal of
Nutrition 102, 1154–1160.
De Vadder, F., Kovatcheva-Datchary, P., Zitoun, C., Duchampt, A., Bäckhed, F., Mithieux, G.,
2016. Microbiota-produced succinate improves glucose homeostasis via intestinal glu-
coneogenesis. Cell Metabolism 24, 151–157.
Delzenne, N.M., Williams, C.M., 2002. Prebiotics and lipid metabolism. Current Opinion in
Lipidology 13, 61–67.
Dietz, W., Santos-Burgoa, C., 2020. Obesity and its implications for COVID-19 mortality.
Obesity (Silver Spring) 28, 1005.
Everard, A., Lazarevic, V., Derrien, M., Girard, M., Muccioli, G.G., Neyrinck, A.M.,
Possemiers, S., Van Holle, A., François, P., de Vos, W.M., 2011. Responses of gut micro-
biota and glucose and lipid metabolism to prebiotics in genetic obese and diet-induced
leptin-resistant mice. Diabetes 60, 2775–2786.
Fan, Y., Pedersen, O., 2021. Gut microbiota in human metabolic health and disease. Nature
Reviews Microbiology 19, 55–71.
Fava, F., Gitau, R., Griffin, B.A., Gibson, G., Tuohy, K., Lovegrove, J., 2013. The type and
quantity of dietary fat and carbohydrate alter faecal microbiome and short-chain fatty
acid excretion in a metabolic syndrome ‘at-risk’population. International Journal of
Obesity 37, 216–223.
Ferretti, P., Pasolli, E., Tett, A., Asnicar, F., Gorfer, V., Fedi, S., Armanini, F., Truong, D.T.,
Manara, S., Zolfo, M., 2018. Mother-to-infant microbial transmission from different body
sites shapes the developing infant gut microbiome. Cell Host & Microbe 24, 133–145.
Fetissov, S.O., Sinno, M.H., Coëffier, M., Bole-Feysot, C., Ducrotté, P., Hökfelt, T., Déchelotte,
P., 2008. Autoantibodies against appetite-regulating peptide hormones and neuropep-
tides: putative modulation by gut microflora. Nutrition 24, 348–359.
Flint, H.J., Duncan, S.H., Scott, K.P., Louis, P., 2015. Links between diet, gut microbiota com-
position and gut metabolism. Proceedings of the Nutrition Society 74, 13–22.
Foster, J.A., Neufeld, K.-A.M., 2013. Gut–brain axis: how the microbiome influences anxiety
and depression. Trends in Neurosciences 36, 305–312.
Francavilla, R., Calasso, M., Calace, L., Siragusa, S., Ndagijimana, M., Vernocchi, P.,
Brunetti, L., Mancino, G., Tedeschi, G., Guerzoni, E., 2012. Effect of lactose on gut
microbiota and metabolome of infants with cow’s milk allergy. Pediatric Allergy and
Immunology 23, 420–427.
Gao, Z., Yin, J., Zhang, J., Ward, R.E., Martin, R.J., Lefevre, M., Cefalu, W.T., Ye, J., 2009.
Butyrate improves insulin sensitivity and increases energy expenditure in mice. Diabetes
58, 1509–1517.
Garcia-Mantrana, I., Selma-Royo, M., Alcantara, C., Collado, M.C., 2018. Shifts on gut
microbiota associated to mediterranean diet adherence and specific dietary intakes on
general adult population. Frontiers in Microbiology 9, 890.
Glick-Bauer, M., Yeh, M.-C., 2014. The health advantage of a vegan diet: exploring the gut
microbiota connection. Nutrients 6, 4822–4838.
Gonze, D., Coyte, K.Z., Lahti, L., Faust, K., 2018. Microbial communities as dynamical
systems. Current Opinion in Microbiology 44, 41–49.
Gribble, F.M., Reimann, F., 2016. Enteroendocrine cells: chemosensors in the intestinal
epithelium. Annual review of physiology 78, 277–299.
Gupta, S., Allen-Vercoe, E., Petrof, E.O., 2016. Fecal microbiota transplantation: in perspec-
tive. Therapeutic Advances in Gastroenterology 9, 229–239.
Diet, Obesity, and Gut Microbiome 233
Han, K., Bose, S., Wang, J., Kim, B.S., Kim, M.J., Kim, E.J., Kim, H., 2015. Contrasting
effects of fresh and fermented kimchi consumption on gut microbiota composition and
gene expression related to metabolic syndrome in obese Korean women. Molecular
Nutrition & Food Research 59, 1004–1008.
Heiss, C.N., Mannerås-Holm, L., Lee, Y.S., Serrano-Lobo, J., Gladh, A.H., Seeley, R.J.,
Drucker, D.J., Bäckhed, F., Olofsson, L.E., 2021. The gut microbiota regulates hypo-
thalamic inflammation and leptin sensitivity in Western diet-fed mice via a GLP-1R-
dependent mechanism. Cell Reports 35, 109163.
Heiss, C.N., Olofsson, L.E., 2018. Gut microbiota-dependent modulation of energy metabo-
lism. Journal of Innate Immunity 10, 163–171.
Hibberd, M.C., Wu, M., Rodionov, D.A., Li, X., Cheng, J., Griffin, N.W., Barratt, M.J.,
Giannone, R.J., Hettich, R.L., Osterman, A.L., 2017. The effects of micronutrient defi-
ciencies on bacterial species from the human gut microbiota. Science Translational
Medicine 9, eaal4069.
Holmes, E., Li, J.V., Marchesi, J.R., Nicholson, J.K., 2012. Gut microbiota composition and activ-
ity in relation to host metabolic phenotype and disease risk. Cell Metabolism 16, 559–564.
Hume, M.P., Nicolucci, A.C., Reimer, R.A., 2017. Prebiotic supplementation improves appe-
tite control in children with overweight and obesity: a randomized controlled trial.
The American Journal of Clinical Nutrition 105, 790–799.
Hussain, M.M., Pan, X., 2009. Clock genes, intestinal transport and plasma lipid homeostasis.
Trends in Endocrinology & Metabolism 20, 177–185.
Ivey, K.L., Hodgson, J.M., Kerr, D.A., Lewis, J.R., Thompson, P.L., Prince, R.L., 2014.
The effects of probiotic bacteria on glycaemic control in overweight men and women:
a randomised controlled trial. European Journal of Clinical Nutrition 68, 447–452.
Jumpertz, R., Le, D.S., Turnbaugh, P.J., Trinidad, C., Bogardus, C., Gordon, J.I., Krakoff, J.,
2011. Energy-balance studies reveal associations between gut microbes, caloric load, and
nutrient absorption in humans. The American Journal of Clinical Nutrition 94, 58–65.
Juttukonda, L.J., Berends, E.T., Zackular, J.P., Moore, J.L., Stier, M.T., Zhang, Y., Schmitz,
J.E., Beavers, W.N., Wijers, C.D., Gilston, B.A., 2017. Dietary manganese promotes
staphylococcal infection of the heart. Cell Host & Microbe 22, 531–542.
Kaakoush, N.O., Day, A.S., Huinao, K.D., Leach, S.T., Lemberg, D.A., Dowd, S.E.,
Mitchell, H.M., 2012. Microbial dysbiosis in pediatric patients with Crohn’s disease.
Journal of Clinical Microbiology 50, 3258–3266.
Kadooka, Y., Sato, M., Imaizumi, K., Ogawa, A., Ikuyama, K., Akai, Y., Okano, M.,
Kagoshima, M., Tsuchida, T., 2010. Regulation of abdominal adiposity by probiotics
(Lactobacillus gasseri SBT2055) in adults with obese tendencies in a randomized con-
trolled trial. European Journal of Clinical Nutrition 64, 636–643.
Kamada, N., Seo, S.-U., Chen, G.Y., Núñez, G., 2013. Role of the gut microbiota in immunity
and inflammatory disease. Nature Reviews Immunology 13, 321–335.
Kim, B., Choi, H.-N., Yim, J.-E., 2019. Effect of diet on the gut microbiota associated with
obesity. Journal of Obesity & Metabolic Syndrome 28, 216.
Kimura, I., Ozawa, K., Inoue, D., Imamura, T., Kimura, K., Maeda, T., Terasawa, K.,
Kashihara, D., Hirano, K., Tani, T., 2013. The gut microbiota suppresses insulin-mediated
fat accumulation via the short-chain fatty acid receptor GPR43. Nature Communications
4, 1–12.
Kocełak, P., Zak-Gołąb, A., Zahorska-Markiewicz, B., Aptekorz, M., Zientara, M.,
Martirosian, G., Chudek, J., Olszanecka-Glinianowicz, M., 2013. Resting energy expen-
diture and gut microbiota in obese and normal weight subjects. European Review for
Medical and Pharmacological Sciences 17, 2816–2821.
Kojima, M., Kangawa, K., 2005. Ghrelin: structure and function. Physiological Reviews 85,
495–522.
234 The Gut Microbiome: Bench to Table
la Cuesta-Zuluaga, D., Mueller, N.T., Álvarez-Quintero, R., Velásquez-Mejía, E.P., Sierra, J.A.,
Corrales-Agudelo, V., Carmona, J.A., Abad, J.M., Escobar, J.S., 2019. Higher fecal
short-chain fatty acid levels are associated with gut microbiome dysbiosis, obesity,
hypertension and cardiometabolic disease risk factors. Nutrients 11, 51.
Larraufie, P., Martin-Gallausiaux, C., Lapaque, N., Dore, J., Gribble, F., Reimann, F.,
Blottiere, H., 2018. SCFAs strongly stimulate PYY production in human enteroendo-
crine cells. Scientific Reports 8, 1–9.
Lazar, V., Ditu, L.-M., Pircalabioru, G.G., Picu, A., Petcu, L., Cucu, N., Chifiriuc, M.C., 2019.
Gut microbiota, host organism, and diet trialogue in diabetes and obesity. Frontiers in
Nutrition 6, 21.
Le Chatelier, E., Nielsen, T., Qin, J., Prifti, E., Hildebrand, F., Falony, G., Almeida, M.,
Arumugam, M., Batto, J.-M., Kennedy, S., 2013. Richness of human gut microbiome
correlates with metabolic markers. Nature 500, 541–546.
Leeuwendaal, N.K., Cryan, J.F., Schellekens, H., 2021. Gut peptides and the microbiome:
focus on ghrelin. Current Opinion in Endocrinology, Diabetes, and Obesity 28, 243.
Ley, R.E., Turnbaugh, P.J., Klein, S., Gordon, J.I., 2006. Human gut microbes associated with
obesity. Nature 444, 1022–1023.
Liang, X., Bushman, F.D., FitzGerald, G.A., 2015. Rhythmicity of the intestinal microbiota is
regulated by gender and the host circadian clock. Proceedings of the National Academy
of Sciences 112, 10479–10484.
Liang, X., FitzGerald, G.A., 2017. Timing the microbes: the circadian rhythm of the gut
microbiome. Journal of Biological Rhythms 32, 505–515.
Liu, C.M., Kanoski, S.E., 2018. Homeostatic and non-homeostatic controls of feeding behav-
ior: distinct vs. common neural systems. Physiology & Behavior 193, 223–231.
Luczynski, P., McVey Neufeld, K.-A., Oriach, C.S., Clarke, G., Dinan, T.G., Cryan, J.F., 2016.
Growing up in a bubble: using germ-free animals to assess the influence of the gut micro-
biota on brain and behavior. International Journal of Neuropsychopharmacology 19, 1–17.
Luthold, R.V., Fernandes, G.R., Franco-de-Moraes, A.C., Folchetti, L.G., Ferreira, S.R.G.,
2017. Gut microbiota interactions with the immunomodulatory role of vitamin D in
normal individuals. Metabolism 69, 76–86.
Machiels, K., Joossens, M., Sabino, J., De Preter, V., Arijs, I., Eeckhaut, V., Ballet, V., Claes, K.,
Van Immerseel, F., Verbeke, K., 2014. A decrease of the butyrate-producing species
Roseburia hominis and Faecalibacterium prausnitzii defines dysbiosis in patients with
ulcerative colitis. Gut 63, 1275–1283.
Makki, K., Deehan, E.C., Walter, J., Bäckhed, F., 2018. The impact of dietary fiber on gut
microbiota in host health and disease. Cell Host & Microbe 23, 705–715.
Manco, M., Putignani, L., Bottazzo, G.F., 2010. Gut microbiota, lipopolysaccharides, and
innate immunity in the pathogenesis of obesity and cardiovascular risk. Endocrine
Reviews 31, 817–844.
Mariat, D., Firmesse, O., Levenez, F., Guimarăes, V., Sokol, H., Doré, J., Corthier, G., Furet, J.,
2009. The Firmicutes/Bacteroidetes ratio of the human microbiota changes with age.
BMC Microbiology 9, 1–6.
Markowiak, P., Śliżewska, K., 2017. Effects of probiotics, prebiotics, and synbiotics on human
health. Nutrients 9, 1021.
Martin, F.P.J., Dumas, M.E., Wang, Y., Legido-Quigley, C., Yap, I.K., Tang, H., Zirah, S., Murphy,
G.M., Cloarec, O., Lindon, J.C., 2007. A top-down systems biology view of microbiome-
mammalian metabolic interactions in a mouse model. Molecular Systems Biology 3, 112.
McCarville, J.L., Chen, G.Y., Cuevas, V.D., Troha, K., Ayres, J.S., 2020. Microbiota metabo-
lites in health and disease. Annual Review of Immunology 38, 147–170.
Méndez-Díaz, M., Rueda-Orozco, P.E., Ruiz-Contreras, A.E., Prospéro-García, O., 2012.
The endocannabinoid system modulates the valence of the emotion associated to food
ingestion. Addiction Biology 17, 725–735.
Diet, Obesity, and Gut Microbiome 235
Milani, C., Duranti, S., Bottacini, F., Casey, E., Turroni, F., Mahony, J., Belzer, C., Delgado
Palacio, S., Arboleya Montes, S., Mancabelli, L., 2017. The first microbial colonizers of
the human gut: composition, activities, and health implications of the infant gut micro-
biota. Microbiology and Molecular Biology Reviews 81, e00036-00017.
Million, M., Maraninchi, M., Henry, M., Armougom, F., Richet, H., Carrieri, P., Valero, R.,
Raccah, D., Vialettes, B., Raoult, D., 2012. Obesity-associated gut microbiota is enriched
in Lactobacillus reuteri and depleted in bifidobacterium animalis and Methano-
brevibacter smithii. International Journal of Obesity 36, 817–825.
Minaya, D.M., Turlej, A., Joshi, A., Nagy, T., Weinstein, N., DiLorenzo, P., Hajnal, A., Czaja, K.,
2020. Consumption of a high energy density diet triggers microbiota dysbiosis, hepatic
lipidosis, and microglia activation in the nucleus of the solitary tract in rats. Nutrition &
Diabetes 10, 1–12.
Moreno-Indias, I., Sánchez-Alcoholado, L., Pérez-Martínez, P., Andrés-Lacueva, C.,
Cardona, F., Tinahones, F., Queipo-Ortuño, M.I., 2016. Red wine polyphenols modu-
late fecal microbiota and reduce markers of the metabolic syndrome in obese patients.
Food & function 7, 1775–1787.
Morrison, C.D., Morton, G.J., Niswender, K.D., Gelling, R.W., Schwartz, M.W., 2005.
Leptin inhibits hypothalamic Npy and Agrp gene expression via a mechanism that
requires phosphatidylinositol 3-OH-kinase signaling. American Journal of Physiology-
Endocrinology and Metabolism 289, E1051–E1057.
Morton, G., Schwartz, M., 2001. The NPY/AgRP neuron and energy homeostasis. International
Journal of Obesity 25, S56–S62.
Mu, C., Yang, Y., Zhu, W., 2016. Gut microbiota: the brain peacekeeper. Frontiers in
Microbiology 7, 345.
Nejrup, R.G., Licht, T.R., Hellgren, L.I., 2017. Fatty acid composition and phospholipid types
used in infant formulas modifies the establishment of human gut bacteria in germ-free
mice. Scientific Reports 7, 1–11.
Ng, S.C., Xu, Z., Mak, J.W.Y., Yang, K., Liu, Q., Zuo, T., Tang, W., Lau, L., Lui, R.N.,
Wong, S.H., 2021. Microbiota engraftment after faecal microbiota transplantation in
obese subjects with type 2 diabetes: a 24-week, double-blind, randomised controlled
trial. Gut 71, 716–723.
O’Toole, P.W., Jeffery, I.B., 2015. Gut microbiota and aging. Science 350, 1214–1215.
Osterberg, K.L., Boutagy, N.E., McMillan, R.P., Stevens, J.R., Frisard, M.I., Kavanaugh, J.W.,
Davy, B.M., Davy, K.P., Hulver, M.W., 2015. Probiotic supplementation attenuates
increases in body mass and fat mass during high-fat diet in healthy young adults.
Obesity 23, 2364–2370.
Parnell, J.A., Reimer, R.A., 2009. Weight loss during oligofructose supplementation is associ-
ated with decreased ghrelin and increased peptide YY in overweight and obese adults.
The American Journal of Clinical Nutrition 89, 1751–1759.
Pedret, A., Valls, R.M., Calderón-Pérez, L., Llauradó, E., Companys, J., Pla-Pagà, L.,
Moragas, A., Martín-Luján, F., Ortega, Y., Giralt, M., 2019. Effects of daily consump-
tion of the probiotic Bifidobacterium animalis subsp. lactis CECT 8145 on anthropo-
metric adiposity biomarkers in abdominally obese subjects: a randomized controlled
trial. International Journal of Obesity 43, 1863–1868.
Perry, R.J., Peng, L., Barry, N.A., Cline, G.W., Zhang, D., Cardone, R.L., Petersen, K.F.,
Kibbey, R.G., Goodman, A.L., Shulman, G.I., 2016. Acetate mediates a microbiome–
brain–β-cell axis to promote metabolic syndrome. Nature 534, 213–217.
Pineiro, M., Asp, N.-G., Reid, G., Macfarlane, S., Morelli, L., Brunser, O., Tuohy, K.,
2008. FAO Technical meeting on prebiotics. Journal of Clinical Gastroenterology
42, S156–S159.
Plaza-Diaz, J., Ruiz-Ojeda, F.J., Gil-Campos, M., Gil, A., 2019. Mechanisms of action of
probiotics. Advances in Nutrition 10, S49–S66.
236 The Gut Microbiome: Bench to Table
Popkin, B.M., Du, S., Green, W.D., Beck, M.A., Algaith, T., Herbst, C.H., Alsukait, R.F.,
Alluhidan, M., Alazemi, N., Shekar, M., 2020. Individuals with obesity and COVID-19:
a global perspective on the epidemiology and biological relationships. Obesity Reviews
21, e13128.
Queipo-Ortuño, M.I., Boto-Ordóñez, M., Murri, M., Gomez-Zumaquero, J.M., Clemente-
Postigo, M., Estruch, R., Cardona Diaz, F., Andres-Lacueva, C., Tinahones, F.J., 2012.
Influence of red wine polyphenols and ethanol on the gut microbiota ecology and bio-
chemical biomarkers. The American Journal of Clinical Nutrition 95, 1323–1334.
Queipo-Ortuño, M.I., Seoane, L.M., Murri, M., Pardo, M., Gomez-Zumaquero, J.M.,
Cardona, F., Casanueva, F., Tinahones, F.J., 2013. Gut microbiota composition in male
rat models under different nutritional status and physical activity and its association
with serum leptin and ghrelin levels. PLoS One 8, e65465.
Rao, C., Coyte, K.Z., Bainter, W., Geha, R.S., Martin, C.R., Rakoff-Nahoum, S., 2021. Multi-
kingdom ecological drivers of microbiota assembly in preterm infants. Nature 591,
633–638.
Rau, A.R., Hentges, S.T., 2019. GABAergic inputs to POMC neurons originating from the
dorsomedial hypothalamus are regulated by energy state. The Journal of Neuroscience
39, 6449–6459.
Reddel, S., Putignani, L., Del Chierico, F., 2019. The impact of low-FODMAPs, gluten-free, and
ketogenic diets on gut microbiota modulation in pathological conditions. Nutrients 11, 373.
Renz, H., Skevaki, C., 2021. Early life microbial exposures and allergy risks: opportunities for
prevention. Nature Reviews Immunology 21, 177–191.
Rinninella, E., Raoul, P., Cintoni, M., Franceschi, F., Miggiano, G.A.D., Gasbarrini, A.,
Mele, M.C., 2019. What is the healthy gut microbiota composition? A changing eco-
system across age, environment, diet, and diseases. Microorganisms 7, 14.
Rooks, M.G., Garrett, W.S., 2016. Gut microbiota, metabolites and host immunity. Nature
Reviews Immunology 16, 341–352.
Rosenbaum, M., Knight, R., Leibel, R.L., 2015. The gut microbiota in human energy homeo-
stasis and obesity. Trends in Endocrinology & Metabolism 26, 493–501.
Russell, W.R., Gratz, S.W., Duncan, S.H., Holtrop, G., Ince, J., Scobbie, L., Duncan, G.,
Johnstone, A.M., Lobley, G.E., Wallace, R.J., 2011. High-protein, reduced-carbohydrate
weight-loss diets promote metabolite profiles likely to be detrimental to colonic health.
The American Journal of Clinical Nutrition 93, 1062–1072.
Sáez-Lara, M.J., Robles-Sanchez, C., Ruiz-Ojeda, F.J., Plaza-Diaz, J., Gil, A., 2016. Effects
of probiotics and synbiotics on obesity, insulin resistance syndrome, type 2 diabetes
and non-alcoholic fatty liver disease: a review of human clinical trials. International
Journal of Molecular Sciences 17, 928.
Sánchez, B., Delgado, S., Blanco‐Míguez, A., Lourenço, A., Gueimonde, M., Margolles, A., 2017.
Probiotics, gut microbiota, and their influence on host health and disease. Molecular
Nutrition & Food Research 61, 1600240.
Scheiman, J., Luber, J.M., Chavkin, T.A., MacDonald, T., Tung, A., Pham, L.D., Wibowo, M.C.,
Wurth, R.C., Punthambaker, S., Tierney, B.T., Yang, Z., Hattab, M.W., Avila-Pacheco, J.,
Clish, C.B., Lessard, S., Church, G.M., Kostic, A.D., 2019. Meta-omics analysis of elite
athletes identifies a performance-enhancing microbe that functions via lactate metabo-
lism. Nature Medicine 25, 1104–1109.
Schéle, E., Grahnemo, L., Anesten, F., Hallén, A., Bäckhed, F., Jansson, J.-O., 2013. The gut
microbiota reduces leptin sensitivity and the expression of the obesity-suppressing
neuropeptides proglucagon (Gcg) and brain-derived neurotrophic factor (Bdnf) in the
central nervous system. Endocrinology 154, 3643–3651.
Sen, T., Cawthon, C.R., Ihde, B.T., Hajnal, A., DiLorenzo, P.M., Claire, B., Czaja, K., 2017.
Diet-driven microbiota dysbiosis is associated with vagal remodeling and obesity.
Physiology & Behavior 173, 305–317.
Diet, Obesity, and Gut Microbiome 237
Sharafedtinov, K.K., Plotnikova, O.A., Alexeeva, R.I., Sentsova, T.B., Songisepp, E.,
Stsepetova, J., Smidt, I., Mikelsaar, M., 2013. Hypocaloric diet supplemented with pro-
biotic cheese improves body mass index and blood pressure indices of obese hyper-
tensive patients-a randomized double-blind placebo-controlled pilot study. Nutrition
Journal 12, 1–11.
Singh, R.K., Chang, H.-W., Yan, D., Lee, K.M., Ucmak, D., Wong, K., Abrouk, M., Farahnik, B.,
Nakamura, M., Zhu, T.H., 2017. Influence of diet on the gut microbiome and implica-
tions for human health. Journal of Translational Medicine 15, 1–17.
Smits, L.P., Bouter, K.E., de Vos, W.M., Borody, T.J., Nieuwdorp, M., 2013. Therapeutic
potential of fecal microbiota transplantation. Gastroenterology 145, 946–953.
Sobhani, I., Tap, J., Roudot-Thoraval, F., Roperch, J.P., Letulle, S., Langella, P., Corthier,
G., Van Nhieu, J.T., Furet, J.P., 2011. Microbial dysbiosis in colorectal cancer (CRC)
patients. PLoS One 6, e16393.
Sonnenburg, E.D., Smits, S.A., Tikhonov, M., Higginbottom, S.K., Wingreen, N.S.,
Sonnenburg, J.L., 2016. Diet-induced extinctions in the gut microbiota compound over
generations. Nature 529, 212–215.
Statovci, D., Aguilera, M., MacSharry, J., Melgar, S., 2017. The impact of western diet and
nutrients on the microbiota and immune response at mucosal interfaces. Frontiers in
Immunology 8, 838.
Stiemsma, L.T., Nakamura, R.E., Nguyen, J.G., Michels, K.B., 2020. Does consumption of fer-
mented foods modify the human gut microbiota? The Journal of nutrition 150, 1680–1692.
Suárez-Zamorano, N., Fabbiano, S., Chevalier, C., Stojanović, O., Colin, D.J., Stevanović, A.,
Veyrat-Durebex, C., Tarallo, V., Rigo, D., Germain, S., 2015. Microbiota depletion
promotes browning of white adipose tissue and reduces obesity. Nature Medicine 21,
1497–1501.
Suez, J., Korem, T., Zeevi, D., Zilberman-Schapira, G., Thaiss, C.A., Maza, O., Israeli, D.,
Zmora, N., Gilad, S., Weinberger, A., 2014. Artificial sweeteners induce glucose intol-
erance by altering the gut microbiota. Nature 514, 181–186.
Sun, L., Ma, L., Ma, Y., Zhang, F., Zhao, C., Nie, Y., 2018. Insights into the role of gut micro-
biota in obesity: pathogenesis, mechanisms, and therapeutic perspectives. Protein &
Cell 9, 397–403.
Symonds, E.L., Peiris, M., Page, A.J., Chia, B., Dogra, H., Masding, A., Galanakis, V., Atiba, M.,
Bulmer, D., Young, R.L., 2015. Mechanisms of activation of mouse and human entero-
endocrine cells by nutrients. Gut 64, 618–626.
Thaiss, C.A., Zeevi, D., Levy, M., Zilberman-Schapira, G., Suez, J., Tengeler, A.C., Abramson, L.,
Katz, M.N., Korem, T., Zmora, N., 2014. Transkingdom control of microbiota diurnal
oscillations promotes metabolic homeostasis. Cell 159, 514–529.
Thursby, E., Juge, N., 2017. Introduction to the human gut microbiota. Biochemical Journal
474, 1823–1836.
Tirosh, A., Calay, E.S., Tuncman, G., Claiborn, K.C., Inouye, K.E., Eguchi, K., Alcala, M.,
Rathaus, M., Hollander, K.S., Ron, I., 2019. The short-chain fatty acid propionate
increases glucagon and FABP4 production, impairing insulin action in mice and
humans. Science Translational Medicine 11, eaav0120.
Tolhurst, G., Heffron, H., Lam, Y.S., Parker, H.E., Habib, A.M., Diakogiannaki, E., Cameron, J.,
Grosse, J., Reimann, F., Gribble, F.M., 2012. Short-chain fatty acids stimulate gluca-
gon-like peptide-1 secretion via the G-protein–coupled receptor FFAR2. Diabetes 61,
364–371.
Topping, D.L., Clifton, P.M., 2001. Short-chain fatty acids and human colonic function:
roles of resistant starch and nonstarch polysaccharides. Physiological Reviews. 81,
1031–1064.
Torres-Fuentes, C., Schellekens, H., Dinan, T.G., Cryan, J.F., 2017. The microbiota–gut–brain
axis in obesity. The Lancet Gastroenterology & Hepatology 2, 747–756.
238 The Gut Microbiome: Bench to Table
Turnbaugh, P.J., Hamady, M., Yatsunenko, T., Cantarel, B.L., Duncan, A., Ley, R.E., Sogin, M.L.,
Jones, W.J., Roe, B.A., Affourtit, J.P., 2009. A core gut microbiome in obese and lean
twins. Nature 457, 480–484.
Turnbaugh, P.J., Ley, R.E., Mahowald, M.A., Magrini, V., Mardis, E.R., Gordon, J.I., 2006.
An obesity-associated gut microbiome with increased capacity for energy harvest.
Nature 444, 1027–1031.
Uzbay, T., 2019. Germ-free animal experiments in the gut microbiota studies. Current Opinion
in Pharmacology 49, 6–10.
Vaiserman, A.M., Koliada, A.K., Marotta, F., 2017. Gut microbiota: a player in aging and a
target for anti-aging intervention. Ageing Research Reviews 35, 36–45.
van Son, J., Koekkoek, L.L., La Fleur, S.E., Serlie, M.J., Nieuwdorp, M., 2021. The role of the
gut microbiota in the gut–brain axis in obesity: mechanisms and Future Implications.
International Journal of Molecular Sciences 22, 2993.
Vaughn, A.C., Cooper, E.M., DiLorenzo, P.M., O’Loughlin, L.J., Konkel, M.E., Peters, J.H.,
Hajnal, A., Sen, T., Lee, S.H., de La Serre, C.B., 2017. Energy-dense diet triggers
changes in gut microbiota, reorganization of gut-brain vagal communication and
increases body fat accumulation. Acta Neurobiologiae Experimentalis 77, 18.
Voigt, R., Forsyth, C., Green, S., Engen, P., Keshavarzian, A., 2016. Circadian rhythm and the
gut microbiome. International Review of Neurobiology 131, 193–205.
Vrieze, A., Van Nood, E., Holleman, F., Salojärvi, J., Kootte, R.S., Bartelsman, J.F., Dallinga–
Thie, G.M., Ackermans, M.T., Serlie, M.J., Oozeer, R., 2012. Transfer of intestinal
microbiota from lean donors increases insulin sensitivity in individuals with metabolic
syndrome. Gastroenterology 143, 913–916.
Willemsen, L., Koetsier, M., Van Deventer, S., Van Tol, E., 2003. Short chain fatty acids
stimulate epithelial mucin 2 expression through differential effects on prostaglandin E1
and E2 production by intestinal myofibroblasts. Gut 52, 1442–1447.
World Health Organization, 2020. "Overweight and obesity", in Health at a Glance: Asia/
Pacific 2020: Measuring Progress Towards Universal Health Coverage, OECD
Publishing, Paris, https://doi.org/10.1787/a47d0cd2-en.
Wright, H., Li, X., Fallon, N.B., Crookall, R., Giesbrecht, T., Thomas, A., Halford, J.C.,
Harrold, J., Stancak, A., 2016. Differential effects of hunger and satiety on insular cor-
tex and hypothalamic functional connectivity. European Journal of Neuroscience 43,
1181–1189.
Xuan, C., Shamonki, J.M., Chung, A., DiNome, M.L., Chung, M., Sieling, P.A., Lee, D.J.,
2014. Microbial dysbiosis is associated with human breast cancer. PLoS One 9, e83744.
Yilmaz, B., Li, H., 2018. Gut microbiota and iron: the crucial actors in health and disease.
Pharmaceuticals 11, 98.
Yu, K.B., Hsiao, E.Y., 2021. Roles for the gut microbiota in regulating neuronal feeding cir-
cuits. Journal of Clinical Investigation 131, e143772.
Zhao, L., 2013. The gut microbiota and obesity: from correlation to causality. Nature Reviews
Microbiology 11, 639–647.
Zhao, L., Zhang, F., Ding, X., Wu, G., Lam, Y.Y., Wang, X., Fu, H., Xue, X., Lu, C., Ma, J., 2018.
Gut bacteria selectively promoted by dietary fibers alleviate type 2 diabetes. Science 359,
1151–1156.
Zhou, Y., Rui, L., 2013. Leptin signaling and leptin resistance. Frontiers of Medicine 7,
207–222.
Zmora, N., Suez, J., Elinav, E., 2019. You are what you eat: diet, health and the gut microbiota.
Nature Reviews Gastroenterology Hepatology 16, 35–56.
9 Challenge, Future
Research, and Influence
in Food Science and
Product Development
Marta Vernero
University of Pavia
CONTENTS
Food and Gut Microbiome ..................................................................................... 239
Microbiota Investigation Tools: Present and Future ..............................................240
Culturomics: A New Approach to Investigate Human Gut Microbiota ................. 242
Role of Functional Food in Microbiota Modulation .............................................. 243
The Journey of Microbiome through Human Evolution ........................................ 245
References ..............................................................................................................248
fat), especially if associated with high sugar and low fiber intake, increases bile-toler-
ant microorganisms including Bacteroides, Bilophila, Alistipes, and Ruminococcus,
decreasing those polysaccharides digesting species such as Firmicutes (Mukhopadhya
et al. 2012; David et al. 2014). Children living in Burkina Faso have a prevalence
of Bacteroidetes in their microbiome, while Italian children, with opposite dietary
habit, have a majority of Enterobacteriaceae.
This finding suggests that carbohydrate and fiber intake is the key to richness in gut
microbiota composition (Yatsunenko et al. 2012). Fibers divide into complex carbohy-
drates and oligosaccharides with different impacts on the gut. Complex carbohydrates
usually contain a huge part of indigestible material that is eliminated through stools,
called starch. These indigestible fibers can be divided into four subgroups, depending
on the resistance features: type 1 is characterized by plant cell wall polymers, type
2 is characterized by a granular structure, type 3 is derived from retro gradiation by
means of heating and coiling, and type 4 is represented by chemical cross-linking
(Flint et al. n.d.). Particularly, resistant starch type 2 increases Ruminococcus spp. and
Eubacterium rectale, whereas type 3 promotes Roseburia spp. and Ruminococcus
bromii (Leitch et al. 2007). Type 4 starch increases Parabacteroides distasonis but
reduces Euacterium rectale and Ruminococcus bromii. This last one also leads to a
reduction of Firmicutes while increasing Bacteroides and Actinobacteria (Martínez
et al. 2010). In addition, a diet high in soluble fibers is usually associated with an
increase in butyrate producing bacteria such as Clostridium leptum and Eubacterium
rectale and of other beneficial bacteria such as Bifidobacterium longum. Particularly,
in vegetarian individuals, the most represented bacteria is Prevotella spp. (Dewulf
et al. 2013). On the contrary, a low-fiber diet significantly decreases all of the afore-
mentioned bacteria (Bibbò et al. 2016). Oligosaccharides, including fructooligosac-
charides, galactooligosaccharides, and arabinoxylan-oligosaccharides, can influence
microbiota composition. For instance, inulin and fructooligosaccharides increase
Bifidobacterium spp. and Lactobacillus spp. and butyrate producing bacteria such as
Faecalibacterium prausnitzii (Ramirez-Farias et al. 2009). On the other hand, galac-
tooligosaccharides usually increases Bifidobacteria spp. (adolescentis and catenula-
tum) and Faecalibacterium prausnitzii, although its effect can be different in each
subject (Ramirez-Farias et al. 2009; Davis et al. 2011).
Additionally, the role of fatty acids in gut microbiota modulation has been exten-
sively studied: A high intake in monounsaturated fatty acids decreases Bifidobacteria
spp. and slightly improves Bacteroides spp. (Simões et al. 2013). In addition to that a
high-fat diet can lead to dysbiosis through reduction of Roseburia species (Neyrinck
et al. 2011).
inhabiting the average human intestine, whereas the majority of microbial species
could never have been cultured, studied, or quantified in a laboratory. Great progress
has been made by the advent of molecular biology, a DNA-based culture-independent
method. By doing this, researchers received a key tool to investigate several aspects
of microbial communities (e.g., taxonomic composition and functional metagenom-
ics) and (theoretically) to deduce potential biological tasks carried out by a commu-
nity as a whole.
Starting from the earliest DNA-based methods by using fluorescent in-situ
hybridization (FISH), passing through polymerase chain reaction (PCR), we
arrived to the revolutionary technology of high-throughput sequencing, known
as next-generation sequencing (NGS). NGS exhibits substantial advances over
the Sanger method in terms of ease and cost of sequencing, as complete bacterial
genome sequences could be assayed and dissected in hours or days rather than
months or years (Hiergeist et al. 2015).
With the development of these methods, two different approaches became avail-
able: the 16S gene analysis and metagenomics. 16S DNA sequencing needs the DNA
extraction from the sample of choice and the use of a specific primer to allow the PCR
to begin. It usually looks for a fundamental part of the set of prokaryotic functional
genes; therefore, only bacteria can be detected. These hypervariable gene regions
could define the genetic fingerprint of a single microorganism. In fact, such regions
have been numbered from V1 to V9 and every one of them encodes specific primers.
To notice, some authors pointed out that there is no hypervariable region that alone
allows the diagnosis of all the bacterial genera examined, so it would be advisable to
investigate different regions (Martellacci et al. 2019).
On the other hand, metagenomics is the study of genetic material recovered
directly from environmental samples, such as fecal samples, in an untargeted (shot-
gun) way without the need to first cultivate these organisms or specific primers.
It relies on a labor-intensive cloning process and on NGS technologies such as
the 454/Roche, Illumina/Solexa, and Ion Torrent/Ion Proton platforms (Garza and
Dutilh 2015).
In particular, Shotgun metagenomics, which “fragments” and then “reads” the
entire DNA in the sample, is able to detect viruses, bacteria, and parasites.
Comparing these two methods is quite clear that metagenomics approach presents
several advantages: (i) it needs less amount of PCR amplification than 16S DNA
sequence analysis; (ii) it is able to quantify individual bacterial species in samples;
and (iii) only with metagenomic approach is possible to identify genetic segments
potentially associated with health or disease. However, it has to be considered that
16S DNA sequencing has lower costs and less computational requirements than
metagenomics, and for these reasons, it is often preferred by researchers (Martellacci
et al. 2019).
Overall, metagenomics revolutionized the understanding of the relations among
the human microbiome, health, and diseases and supplanted the long and labori-
ous culture-dependent methods of classic microbiology (Lagier et al. 2016). With
metagenomic approaches, we can discover the identity, evolution, gene composition,
distribution, and ecological patterns of uncultured microbes and viruses (Garza and
Dutilh 2015). However, it generates a countless number of sequences that have not
242 The Gut Microbiome: Bench to Table
development, immunity, and the susceptibility of a vast range of acute and chronic
diseases (Marchesi and Ravel 2015). With the initiation of the Human Microbiome
Project in 2007, as a direct consequence of the improvement of DNA sequencing, it
has become increasingly clear that the study of human evolution could exclude the
study of human gut microbiome (Peterson et al. 2009).
Gut microbiome and its co-resident microbes contain a huge quantity of different
additional genes, functioning as a target for natural selection (Schnorr et al. 2016).
So, the study of gut microorganisms in human populations, non-human primates,
and past human populations is a key point in understanding human evolution, even
though this is a novel field still lacking strong evidences. In fact, up to date, the only
archaeological materials containing stable components allowing ancient microbiome
sequencing are coprolites (paleofeces) from mummified human rests and dental cal-
culus (Zeng et al. 2015). The majority of these materials come from samples origi-
nated in Europe and Americas.
Interestingly, environmental and lifestyle changes that have occurred through-
out human history and prehistory had a huge impact on microbiome composition
and physiology.
In fact, unlike the nuclear and mitochondrial genomes of the host, the micro-
biome continuously responds to external and internal changes and signaling
(Dewulf et al. 2013).
Particularly, gut microbiome can change through both vertical (parental trans-
mission) and horizontal transmission (more typical of microorganisms, information
derive from the environment) (Davenport et al. 2017).
Moeller et al. investigated the changes in the passage from apes microbiome
to human microbiome and found out that among apes, Prevotella was negatively
correlated with Bacterioides, but Bacterioides were positively correlated with
Ruminococcus and Parabacteroides. Most of these changes in the composition of
the human microbiome have functional implications for host nutrition. For instance,
the abundance of Bacteroides, which has been positively associated with diets rich
in animal fat and protein, has increased in humans. On the other hand, the archaeon
Methanobrevibacter, which promotes the degradation of complex plant polysac-
charides, has reduced within humans, as well as Fibrobacter (Moeller et al. 2014;
Kobayashi, Shinkai, and Koike 2008).
Interestingly, among primates, Old World monkeys and apes have the most similar
microbiome to actual human microbiome, rather than New World primates and lemurs.
In fact, captive primates consume less diverse, lower-fiber diets compared to their wild
counterparts mirroring the gradual transition to low-fiber diets of human evolution and
the contrast of modern Western and non-Western diets (Davenport et al. 2017).
Basing on biogeographic analyses, it is clear that there are some specific micro-
bial taxa depending host genotypes and environment. This is evident in a higher
presence of Prevotella, Catenibacterium, Succinivibrio, and Treponema among both
contemporary and ancient populations following the same lifestyle such as hunting
and gathering or agriculture (Gomez et al. 2016; Yatsunenko et al. 2012; Raul Y. Tito
et al. 2012; Raúl Y. Tito et al. 2008).
Recently, some samples recovered from Mexico provided DNA from numer-
ous human gut microbial symbionts (Tito et al. 2008, 2012), with taxonomic profile
Challenge, Future Research, and Influence 247
So, the evolution of gut microbiome basically was dictated by the environmen-
tal changes (especially food related) that occurred during human evolution. The
most important difference between present microbiome composition and apes is the
decreased variability mainly due to fast lifestyle changes. In addition to that, nowa-
days, composition of microbiome is much more different between different individu-
als, depending on where they live and how they eat (Moeller et al. 2014; Bibbò et al.
2016; Ianiro, Tilg, and Gasbarrini 2016).
REFERENCES
Adler, Christina J., Keith Dobney, Laura S. Weyrich, John Kaidonis, Alan W. Walker, Wolfgang
Haak, Corey J.A. Bradshaw, et al. 2013. “Sequencing Ancient Calcified Dental Plaque
Shows Changes in Oral Microbiota with Dietary Shifts of the Neolithic and Industrial
Revolutions.” Nature Genetics 45 (4): 450–55. https://doi.org/10.1038/ng.2536.
Bekar, Onder, Yusuf Yilmaz, and MacIt Gulten. 2011. “Kefir Improves the Efficacy and
Tolerability of Triple Therapy in Eradicating Helicobacter Pylori.” Journal of Medicinal
Food 14 (4): 344–47. https://doi.org/10.1089/jmf.2010.0099.
Bellini, Massimo, Sara Tonarelli, Attila G. Nagy, Andrea Pancetti, Francesco Costa, Angelo
Ricchiuti, Nicola de Bortoli, Marta Mosca, Santino Marchi, and Alessandra Rossi.
2020. “Low FODMAP Diet: Evidence, Doubts, and Hopes.” Nutrients. MDPI AG.
https://doi.org/10.3390/nu12010148.
Bibbò, S., G. Ianiro, V. Giorgio, F. Scaldaferri, L. Masucci, A. Gasbarrini, and G. Cammarota.
2016. “The Role of Diet on Gut Microbiota Composition.” European Review for Medical
and Pharmacological Sciences 20 (22): 4742–49.
Blaser, Martin J., and Stanley Falkow. 2009. “What Are the Consequences of the Disappearing
Human Microbiota?” Nature Reviews Microbiology 7 (12): 887–94. https://doi.org/
10.1038/nrmicro2245.
Caminero, Alberto, Alexandra R. Herrán, Esther Nistal, Jenifer Pérez-Andrés, Luis
Vaquero, Santiago Vivas, José María G. Ruiz de Morales, Silvia M. Albillos, and Javier
Casqueiro. 2014. “Diversity of the Cultivable Human Gut Microbiome Involved in
Gluten Metabolism: Isolation of Microorganisms with Potential Interest for Coeliac
Disease.” FEMS Microbiology Ecology 88 (2): 309–19. https://doi.org/10.1111/1574-
6941.12295.
Chan, Yee Kwan, Mehrbod Estaki, and Deanna L. Gibson. 2013. “Clinical Consequences
of Diet-Induced Dysbiosis.” Annals of Nutrition & Metabolism 63 (Suppl 2): 28–40.
https://doi.org/10.1159/000354902.
Chifiriuc, Mariana Carmen, Alina Badea Cioaca, and Veronica Lazar. 2011. “In Vitro Assay
of the Antimicrobial Activity of Kephir against Bacterial and Fungal Strains.” Anaerobe
17 (6): 433–35. https://doi.org/10.1016/j.anaerobe.2011.04.020.
Davenport, Emily R., Jon G. Sanders, Se Jin Song, Katherine R. Amato, Andrew G. Clark,
and Rob Knight. 2017. “The Human Microbiome in Evolution.” BMC Biology. BioMed
Central Ltd. https://doi.org/10.1186/s12915-017-0454-7.
David, Lawrence A., Corinne F. Maurice, Rachel N. Carmody, David B. Gootenberg, Julie E.
Button, Benjamin E. Wolfe, Alisha V. Ling, et al. 2014. “Diet Rapidly and Reproducibly
Alters the Human Gut Microbiome.” Nature 505 (7484): 559–63. https://doi.org/10.1038/
nature12820.
Davis, Lauren M.G., Inés Martínez, Jens Walter, Caitlin Goin, and Robert W. Hutkins. 2011.
“Barcoded Pyrosequencing Reveals That Consumption of Galactooligosaccharides
Results in a Highly Specific Bifidogenic Response in Humans.” PLoS One 6 (9). https://
doi.org/10.1371/journal.pone.0025200.
Challenge, Future Research, and Influence 249
Dewulf, Evelyne M., Patrice D. Cani, Sandrine P. Claus, Susana Fuentes, Philippe G.B.
Puylaert, Audrey M. Neyrinck, Laure B. Bindels, et al. 2013. “Insight into the
Prebiotic Concept: Lessons from an Exploratory, Double Blind Intervention Study with
Inulin-Type Fructans in Obese Women.” Gut 62 (8): 1112–21. https://doi.org/10.1136/
gutjnl-2012-303304.
Dimidi, Eirini, Selina Rose Cox, Megan Rossi, and Kevin Whelan. 2019. “Fermented
Foods: Definitions and Characteristics, Impact on the Gut Microbiota and Effects on
Gastrointestinal Health and Disease.” Nutrients. MDPI AG. https://doi.org/10.3390/
nu11081806.
Dubourg, Grégory, Jean Christophe Lagier, Catherine Robert, Fabrice Armougom, Perrine
Hugon, Sarah Metidji, Niokhor Dione, et al. 2014. “Culturomics and Pyrosequencing
Evidence of the Reduction in Gut Microbiota Diversity in Patients with Broad-Spectrum
Antibiotics.” International Journal of Antimicrobial Agents 44 (2): 117–24. https://doi.
org/10.1016/j.ijantimicag.2014.04.020.
Fernandez-Feo, M., G. Wei, G. Blumenkranz, F.E. Dewhirst, D. Schuppan, F.G. Oppenheim,
and E.J. Helmerhorst. 2013. “The Cultivable Human Oral Gluten-Degrading Microbiome
and Its Potential Implications in Coeliac Disease and Gluten Sensitivity.” Clinical
Microbiology and Infection 19 (9). https://doi.org/10.1111/1469-0691.12249.
Flint, Harry J., Karen P. Scott, Sylvia H. Duncan, Petra Louis, and Evelyne Forano. n.d.
“Microbial Degradation of Complex Carbohydrates in the Gut.” Gut Microbes 3 (4):
289–306. Accessed January 24, 2020. https://doi.org/10.4161/gmic.19897.
Fouhy, Fiona, Caitriona M. Guinane, Seamus Hussey, Rebecca Wall, C. Anthony Ryan,
Eugene M. Dempsey, Brendan Murphy, et al. 2012. “High-Throughput Sequencing
Reveals the Incomplete, Short-Term Recovery of Infant Gut Microbiota
Following Parenteral Antibiotic Treatment with Ampicillin and Gentamicin.”
Antimicrobial Agents and Chemotherapy 56 (11): 5811–20. https://doi.org/
10.1128/AAC.00789-12.
Garza, Daniel R., and Bas E. Dutilh. 2015. “From Cultured to Uncultured Genome Sequences:
Metagenomics and Modeling Microbial Ecosystems.” Cellular and Molecular Life
Sciences 72 (22): 4287–308. https://doi.org/10.1007/s00018-015-2004-1.
Gomez, Andres, Klara J. Petrzelkova, Michael B. Burns, Carl J. Yeoman, Katherine R.
Amato, Klara Vlckova, David Modry, et al. 2016. “Gut Microbiome of Coexisting
BaAka Pygmies and Bantu Reflects Gradients of Traditional Subsistence Patterns.” Cell
Reports 14 (9): 2142–53. https://doi.org/10.1016/j.celrep.2016.02.013.
He, T., K. Venema, M. G. Priebe, G. W. Welling, R. J.M. Brummer, and R. J. Vonk. 2008. “The
Role of Colonic Metabolism in Lactose Intolerance.” European Journal of Clinical
Investigation. European Journal of Clinical Investigation. https://doi.org/10.1111/j.
1365-2362.2008.01966.x.
Hiergeist, Andreas, Joachim Gläsner, Udo Reischl, and André Gessner. 2015. “Analyses
of Intestinal Microbiota: Culture versus Sequencing.” ILAR Journal 56 (2): 228–40.
https://doi.org/10.1093/ilar/ilv017.
Ianiro, Gianluca, Herbert Tilg, and Antonio Gasbarrini. 2016. “Antibiotics as Deep Modulators
of Gut Microbiota: Between Good and Evil.” Gut 65 (11): 1906–15. https://doi.org/
10.1136/gutjnl-2016-312297.
Jakobsson, Hedvig E., Cecilia Jernberg, Anders F. Andersson, Maria Sjölund-Karlsson,
Janet K. Jansson, and Lars Engstrand. 2010. “Short-Term Antibiotic Treatment Has
Differing Long-Term Impacts on the Human Throat and Gut Microbiome.” PLoS One
5 (3): e9836. https://doi.org/10.1371/journal.pone.0009836.
Jernberg, Cecilia, Sonja Löfmark, Charlotta Edlund, and Janet K. Jansson. 2010. “Long-Term
Impacts of Antibiotic Exposure on the Human Intestinal Microbiota.” Microbiology
156: 3216–3223. https://doi.org/10.1099/mic.0.040618-0.
250 The Gut Microbiome: Bench to Table
Khoury, Nathalie, Stephany El-Hayek, Omayr Tarras, Marwan El-Sabban, Mirvat El-Sibai,
and Sandra Rizk. 2014. “Kefir Exhibits Anti-Proliferative and pro-Apoptotic Effects on
Colon Adenocarcinoma Cells with No Significant Effects on Cell Migration and Invasion.”
International Journal of Oncology 45 (5): 2117–27. https://doi.org/10.3892/ijo.2014.2635.
Kobayashi, Y., T. Shinkai, and S. Koike. 2008. “Ecological and Physiological Characterization
Shows That Fibrobacter Succinogenes Is Important in Rumen Fiber Digestion - Review.”
Folia Microbiologica 53 (3): 195–200. https://doi.org/10.1007/s12223-008-0024z.
Kwon, Ok Kyoung, Kyung Seop Ahn, Mee Young Lee, So Young Kim, Bo Young Park, Mi
Kyoung Kim, In Young Lee, Sei Ryang Oh, and Hyeong Kyu Lee. 2008. “Inhibitory
Effect of Kefiran on Ovalbumin-Induced Lung Inflammation in a Murine Model of
Asthma.” Archives of Pharmacal Research 31 (12): 1590–96. https://doi.org/10.1007/
s12272-001-2156-4.
Lagier, Jean-Christophe, F. Armougom, M. Million, P. Hugon, I. Pagnier, C. Robert, F.
Bittar, G. Fournous, G. Gimenez, M. Maraninchi, J.-F. Trape, E. V. Koonin, B. la
Scola, D. Raoult. 2012. “Microbial Culturomics: Paradigm Shift in the Human Gut
Microbiome Study.” Clinical Microbiology and Infection : The Official Publication
of the European Society of Clinical Microbiology and Infectious Diseases 18 (12):
1185–93. https://doi.org/10.1111/1469-0691.12023.
Lagier, Jean Christophe, Grégory Dubourg, Matthieu Million, Frédéric Cadoret, Melhem
Bilen, Florence Fenollar, Anthony Levasseur, Jean Marc Rolain, Pierre Edouard
Fournier, and Didier Raoult. 2018. “Culturing the Human Microbiota and Culturomics.”
Nature Reviews Microbiology. Nature Publishing Group. https://doi.org/10.1038/
s41579-018-0041-0.
Lagier, Jean-Christophe, Saber Khelaifia, Maryam Tidjani Alou, Sokhna Ndongo, Niokhor
Dione, Perrine Hugon, Aurelia Caputo, et al. 2016. “Culture of Previously Uncultured
Members of the Human Gut Microbiota by Culturomics.” Nature Microbiology 1
(November): 16203. https://doi.org/10.1038/nmicrobiol.2016.203.
Leitch, E. Carol McWilliam, Alan W. Walker, Sylvia H. Duncan, Grietje Holtrop, and
Harry J. Flint. 2007. “Selective Colonization of Insoluble Substrates by Human Faecal
Bacteria.” Environmental Microbiology 9 (3): 667–79. https://doi.org/10.1111/j.1462-
2920.2006.01186.x.
Leite, Analy Machado de Oliveira, Marco Antônio Lemos Miguel, Raquel Silva Peixoto,
Alexandre Soares Rosado, Joab Trajano Silva, and Vania Margaret Flosi Paschoalin.
2013. “Microbiological, Technological and Therapeutic Properties of Kefir: A Natural
Probiotic Beverage.” Brazilian Journal of Microbiology. Sociedade Brasileira de
Microbiologia. https://doi.org/10.1590/S1517-83822013000200001.
Liu, Je-Ruei, Ming Ju Chen, and Chin Wen Lin. 2005. “Antimutagenic and Antioxidant
Properties of Milk-Kefir and Soymilk-Kefir.” Journal of Agricultural and Food
Chemistry 53 (7): 2467–74. https://doi.org/10.1021/jf048934k.
Liu, Je-Ruei, Sheng-Yao Wang, Ming-Ju Chen, Hsiao-Ling Chen, Pei-Ying Yueh, and Chin-
Wen Lin. 2006. “Hypocholesterolaemic Effects of Milk-Kefir and Soyamilk-Kefir in
Cholesterol-Fed Hamsters.” British Journal of Nutrition 95 (5): 939–46. https://doi.
org/10.1079/bjn20061752.
Marchesi, Julian R., and Jacques Ravel. 2015. “The Vocabulary of Microbiome Research:
A Proposal.” Microbiome 3 (1). https://doi.org/10.1186/s40168-015-0094-5.
Martellacci, Leonardo, Gianluca Quaranta, Romeo Patini, Gaetano Isola, Patrizia Gallenzi,
and Luca Masucci. 2019. “A Literature Review of Metagenomics and Culturomics of
the Peri-Implant Microbiome: Current Evidence and Future Perspectives.” Materials
(Basel, Switzerland) 12 (18). https://doi.org/10.3390/ma12183010.
Challenge, Future Research, and Influence 251
Martínez, Inés, Jaehyoung Kim, Patrick R. Duffy, Vicki L. Schlegel, and Jens Walter. 2010.
“Resistant Starches Types 2 and 4 Have Differential Effects on the Composition of the
Fecal Microbiota in Human Subjects.” PLoS One 5 (11). https://doi.org/10.1371/journal.
pone.0015046.
Moeller, Andrew H., Yingying Li, Eitel Mpoudi Ngole, Steve Ahuka-Mundeke, Elizabeth V.
Lonsdorf, Anne E. Pusey, Martine Peeters, Beatrice H. Hahn, and Howard Ochman.
2014. “Rapid Changes in the Gut Microbiome during Human Evolution.” Proceedings
of the National Academy of Sciences of the United States of America 111 (46):
16431–35. https://doi.org/10.1073/pnas.1419136111.
Mukhopadhya, Indrani, Richard Hansen, Emad M. El-Omar, and Georgina L. Hold. 2012.
“IBD-What Role Do Proteobacteria Play?” Nature Reviews. Gastroenterology &
Hepatology 9 (4): 219–30. https://doi.org/10.1038/nrgastro.2012.14.
Murtaza, Nida, Páraic Ó. Cuív, and Mark Morrison. 2017. “Diet and the Microbiome.”
Gastroenterology Clinics of North America 46 (1): 49–60. https://doi.org/10.1016/j.
gtc.2016.09.005.
Neyrinck, Audrey M., Sam Possemiers, Céline Druart, Tom Van de Wiele, Fabienne De
Backer, Patrice D. Cani, Yvan Larondelle, and Nathalie M. Delzenne. 2011. “Prebiotic
Effects of Wheat Arabinoxylan Related to the Increase in Bifidobacteria, Roseburia and
Bacteroides/Prevotella in Diet-Induced Obese Mice.” PloS One 6 (6): e20944. https://
doi.org/10.1371/journal.pone.0020944.
Obregon-Tito, Alexandra J., Raul Y. Tito, Jessica Metcalf, Krithivasan Sankaranarayanan,
Jose C. Clemente, Luke K. Ursell, Zhenjiang Zech Xu, et al. 2015. “Subsistence Strategies
in Traditional Societies Distinguish Gut Microbiomes.” Nature Communications 6.
https://doi.org/10.1038/ncomms7505.
Peters, Danielle L., Wenju Wang, Xu Zhang, Zhibin Ning, Janice Mayne, and Daniel Figeys.
2019. “Metaproteomic and Metabolomic Approaches for Characterizing the Gut
Microbiome.” Proteomics 19 (16): e1800363. https://doi.org/10.1002/pmic.201800363.
Peterson, Jane, Susan Garges, Maria Giovanni, Pamela McInnes, Lu Wang, Jeffery A. Schloss,
Vivien Bonazzi, et al. 2009. “The NIH Human Microbiome Project.” Genome Research
19 (12): 2317–23. https://doi.org/10.1101/gr.096651.109.
Ramirez-Farias, Carlett, Kathleen Slezak, Zoë Fuller, Alan Duncan, Grietje Holtrop, and
Petra Louis. 2009. “Effect of Inulin on the Human Gut Microbiota: Stimulation of
Bifidobacterium Adolescentis and Faecalibacterium Prausnitzii.” The British Journal
of Nutrition 101 (4): 541–50. https://doi.org/10.1017/S0007114508019880.
Rinninella, Emanuele, Marco Cintoni, Pauline Raoul, Loris Riccardo Lopetuso, Franco
Scaldaferri, Gabriele Pulcini, Giacinto Abele Donato Miggiano, Antonio Gasbarrini, and
Maria Cristina Mele. 2019. “Food Components and Dietary Habits: Keys for a Healthy
Gut Microbiota Composition.” Nutrients. MDPI AG. https://doi.org/10.3390/nu11102393.
Sánchez-Tapia, Mónica, Armando R. Tovar, and Nimbe Torres. 2019. “Diet as Regulator of
Gut Microbiota and Its Role in Health and Disease.” Archives of Medical Research 50
(5): 259–68. https://doi.org/10.1016/j.arcmed.2019.09.004.
Schnorr, Stephanie L., Krithivasan Sankaranarayanan, Cecil M. Lewis, and Christina
Warinner. 2016. “Insights into Human Evolution from Ancient and Contemporary
Microbiome Studies.” Current Opinion in Genetics and Development. Elsevier Ltd.
https://doi.org/10.1016/j.gde.2016.07.003.
Simões, Catarina D., Johanna Maukonen, Jaakko Kaprio, Aila Rissanen, Kirsi H. Pietiläinen,
and Maria Saarela. 2013. “Habitual Dietary Intake Is Associated with Stool Microbiota
Composition in Monozygotic Twins.” The Journal of Nutrition 143 (4): 417–23. https://
doi.org/10.3945/jn.112.166322.
252 The Gut Microbiome: Bench to Table
Slimings, Claudia, and Thomas V. Riley. 2014. “Antibiotics and Hospital-Acquired Clostridium
Difficile Infection: Update of Systematic Review and Meta-Analysis.” The Journal of
Antimicrobial Chemotherapy 69 (4): 881–91. https://doi.org/10.1093/jac/dkt477.
Stilling, Roman M., Seth R. Bordenstein, Timothy G. Dinan, and John F. Cryan. 2014.
“Friends with Social Benefits: Host-Microbe Interactions as a Driver of Brain Evolution
and Development?” Frontiers in Cellular and Infection Microbiology 4 (Oct). https://
doi.org/10.3389/fcimb.2014.00147.
Tito, Raúl Y., Dan Knights, Jessica Metcalf, Alexandra J. Obregon-Tito, Lauren Cleeland,
Fares Najar, Bruce Roe, et al. 2012. “Insights from Characterizing Extinct Human Gut
Microbiomes.” PLoS One 7 (12). https://doi.org/10.1371/journal.pone.0051146.
Tito, Raúl Y., Simone Macmil, Graham Wiley, Fares Najar, Lauren Cleeland, Chunmei
Qu, Ping Wang, et al. 2008. “Phylotyping and Functional Analysis of Two Ancient
Human Microbiomes.” PLoS One 3 (11): e3703–e3703. https://doi.org/10.1371/journal.
pone.0003703.
Vasquez, Alex, Sidney MacDonald Baker, Peter Bennett, Jeffrey S. Bland, Leo Galland,
Robert J. Hedaya, Mark Houston, Mark Hyman, Jay Lombard, and Robert Rountree.
2010. Textbook of Functional Medicine. Example Product Manufacturer.
Yano, Jessica M., Kristie Yu, Gregory P. Donaldson, Gauri G. Shastri, Phoebe Ann, Liang
Ma, Cathryn R. Nagler, Rustem F. Ismagilov, Sarkis K. Mazmanian, and Elaine Y.
Hsiao. 2015. “Indigenous Bacteria from the Gut Microbiota Regulate Host Serotonin
Biosynthesis.” Cell 161 (2): 264–76. https://doi.org/10.1016/j.cell.2015.02.047.
Yatsunenko, Tanya, Federico E. Rey, Mark J. Manary, Indi Trehan, Maria Gloria Dominguez-
Bello, Monica Contreras, Magda Magris, et al. 2012. “Human Gut Microbiome Viewed
across Age and Geography.” Nature. https://doi.org/10.1038/nature11053.
Zeng, Qinglong, Jeet Sukumaran, Steven Wu, and Allen Rodrigo. 2015. “Neutral Models of
Microbiome Evolution.” PLoS Computational Biology 11 (7). https://doi.org/10.1371/
journal.pcbi.1004365.
Zhang, Lu, Ying Huang, Yang Zhou, Timothy Buckley, and Hua H. Wang. 2013. “Antibiotic
Administration Routes Significantly Influence the Levels of Antibiotic Resistance in
Gut Microbiota.” Antimicrobial Agents and Chemotherapy 57 (8): 3659–66. https://doi.
org/10.1128/AAC.00670-13.
Index
Note: Bold page numbers refer to tables; italic page numbers refer to figures.
aberrant microbiome 5 ASC see adult stem cell (ASC)
absorbable short-chain fatty acids 4 ASV see Amplicon Sequence Variant (ASV)
Acetobacteraceae (a-proteo-bacteria) 106 atherosclerosis 90
acidic pH 128, 170 Automated Self-Administered 24-hour Dietary
Acinetobacter baumanii 196 Recalls (ASA24) 83
acquired immunodeficiency syndrome (AIDS) avian microbiomes 107
54, 55 avian models 107
Actinobacteria 74, 105, 163, 190, 214, 229
adaptive immune responses 4 Bacillus
ad-hoc heuristic approach 22 B. anthracis 122
adult stem cell (ASC) 102 B. cereus 122
derived intestinal organoids 102 B. coagulans 171
Adventist Health Study 186 B. subtilis 245
AgRP/NPY neurons 220 bacteria
AhR see aryl hydrocarbon receptor (AhR) and metabolites 4
AIDS see acquired immunodeficiency syndrome phages infect 46
(AIDS) probiotic lactic acid, lipids of 128–129
Akkermansia muciniphila 189, 196, 198, 224 SCFA-producing 227
ALDEx2 22–23, 25 bacteria–gut–host physiology system 213
Alexandrium minutum 139 bacterial cell walls, cholesterol integration into
α-linoleic acid, biosynthetic derivatives of 86 128–129
Allobaculum 144 bacterial CRISPR genes 47
Amazon rainforest 157 bacterial host cells, lysing of 52
amino acid 128 bacterial metabolism of dietary fiber 78
metabolism, gut microbiome and 89–90 bacteriophages (phages) 45
Amplicon Sequence Variant (ASV) 10 Bacteroidaceae 108, 144
amplicon sequencing, full-length 16S rRNA 11 Bacteroides 82, 99
amplification cycles 9 abundance in 83
amyloid-producing Escherichia coli 54 Bacteroides 108, 138, 145, 224, 229
Anaerostipes 162 B. dorei 226
analysis of composition of microbiomes B. fragilis 47
(ANCOM) 23 B. vulgatus 224, 226
analysis of similarities (ANOSIM) 17 Bacteroidetes 5, 74, 80–83, 91, 105, 214, 215,
Analysis of variance (ANOVA) 21 217–219
ANCOM see analysis of composition of Bacteroidetes 53, 108, 111
microbiomes (ANCOM) basal metabolic rate (BMR) 91
Anelloviridae 55, 56 Basic Local Alignment Search Tool (BLAST) 14
animal protein, diet rich in 226 Bayesian multiplicative replacement method 25
anthocyanins 186, 192 BCAAs see branched-chain amino acids
antibiotics 135 (BCAAs)
anti-inflammatory action of resveratrol 195 Benjamini–Hochberg procedure 22
anti-inflammatory amino acid metabolites 89 berries’ polyphenols 194
antimicrobial activity against Candida betaine 131
albicans 244 β-glucan polysaccharide 83
arabinoxylan-oligosaccharides 240 Bifidobacteria 80, 86, 89
aromatic acids 76 dominated microbiome 77
aronia (poly)phenol-rich extract 196 Bifidobacteria 133, 136, 137, 144, 145, 158, 162,
artificial sweeteners 225 164, 170, 171, 243
aryl hydrocarbon receptor (AhR) 89 Bifidobacteriaceae 158
253
254 Index
Bifidobacterium 80, 83, 87, 99, 122, 136, 138, carbohydrate-active enzymes (CAZymes) 79
142, 159, 194, 219, 224, 226, 229 carbohydrate metabolism
B. adolescentis 225 gut microbiome and 79–84
B. animalis 228 complex carbohydrates 82
subsp. lactis CECT 8145 228 nondigestible polysaccharides–fiber
B. breve 144, 191 82–84
B. longum 142, 164 simple carbohydrates 80–82
breve ATCC 15700 130 carbohydrate prebiotics 159
BSH activity in 127 human milk oligosaccharides (HMO)
B. vulgatus 218 159–161, 160
bifidogenic effects 162 inulin 162
bile acids (BAs) 76, 126 oligosaccharides 159
microbial cometabolism of 76 polysaccharides 161–162
bile salt 124 carbohydrates 224–225
cholesterol deposition with conjugated 128 complex 82
conjugation 126 simple 80–82
hydrolyzing enzyme cardiovascular disease (CVD) 73, 135–139, 185
decongesting bile salts using 126–128, 127 carotenoids 189
removal of deconjugated 128 carrier matrix 170
bile salt hydrolase (BSH) activity, in casein 89
Bifidobacterium and Lactobacillus 127 catechins 191
Bilophila wadsworthia 85, 144 Caudovirales 54
bioactive compounds, plant 187 phages 56
bioactive polyphenol-derived compounds 183 CCK 223
biogeography of microbiome 80 CD see celiac disease (CD); Crohn’s disease (CD)
bioinformatics methods 7 CD4+ T cells 89
Biological Observation Matrix (BIOM) format 17 cecum microflora 89
bioprinting 102 celiac disease (CD) 243
droplet-based 102 cell culture 100–101
extrusion-based 102 3D 102
biotransformations, microbial enzymatic 164 cell membranes 129
bird microbiomes 107 cellulose 82
BLAST see Basic Local Alignment Search Tool centered log-ratio (CLR) approach 22
(BLAST) Centers for Disease Control and Prevention 10
blood–brain barrier (BBB) 888 C-glycosides 192
permeability 81 cholecystokinin (CCK) 220
BMI see body mass index (BMI) cholesterol
body mass index (BMI) 75, 212 binding of 130
brainstem 221 deposition of
branched-chain amino acids (BCAAs) 193 with colic acid 128
Bray–Curtis dissimilarities 33 deposition with conjugated bile salt 128
breastfeeding 77 integration into bacterial cell walls 128–129
breast milk 50 cholesterol-lowering effect mechanisms 126
triglycerides 77 cholesterol-lowering probiotic effects,
butyrate-producing Faecalibacterium hypothetical concept of 126
prausnitzii 85 cholesterol-lowering probiotics, biotherapeutic
butyrate-supplemented high-fat diets 84 properties of 121–140
Butyrivibrio crossotus 111 binding of cholesterol 130
cholesterol deposition with conjugated bile
Caco-2 103 salt 128
cell line 101 cholesterol integration into bacterial cell
cells 101 walls 128–129
co-cultures of 101 cholesterol-lowering effect mechanisms 126
Calypso 31–33 decongesting bile salts using bile salt
cancer treatment 138–139 hydrolyzing enzyme 126–128, 127
Candida albicans 244 gut microbiota in human health and disease
antimicrobial activity against 244 prevention 134–135
Index 255
microbiome 4, 71, 72; see also specific types milk proteins 168
aberrant 5 MiSeq 9, 10
altered 5 standard operating procedure 18
bifidobacteria-dominated 77 mobile genetic elements (MGEs) 92
biogeography of 80 molecular ecological network analyses
human fetus 4 (MENA) 28
nutritional environment for 78 monocarboxylate transporters 88
placental 4 monounsaturated fatty acids (MUFAs) 244
role in 4 Monte Carlo sampling 22
through human evolution 245–248 mothur 18
and weight change 90–91 motilin 222
Microbiome Analyst 30, 31 mouse models, Lactobacillus reuteri in 51
microbiome connection, metabolome and 75–76 MPI see milk protein isolate (MPI)
microbiome-derived metabolites 4 MPLasso see Microbial Prior LASSO (MPLasso)
microbiome–diet interactions 78, 79 mucosal layers, phages in 50
microbiome metabolic interactions mucosal microbiome 104
host–gut 76–78 murine models 107–109
microbiome-produced enzymes 89 Mycoplasma pneumoniae, chromosome
microbiome profiling, gene–based marker 5 structure of 8
microbiome studies myeloid differentiation factor 2 (MD2) 86
gene-based marker 5
historical perspective of 6–8 Nanopore, Oxford 11
Microbiome Taxonomic Profiling 14 National Center for Biotechnology Information
microbiome zeitgeist 72 (NCBI) taxonomy 12
microbiota 46, 72 NEC see necrotizing enterocolitis (NEC)
colonic 74 necrotizing enterocolitis (NEC) 166
colonizes 47 Neisseria 74
infant’s intestinal 77–78 Nelder’s algorithm 30
microbiota and host stem, symbiotic relationships neonatal’s ingestion of microorganisms 76
between 77 netagenomic approaches 60
microbiota composition 191 network algorithms 26
microbiota-derived metabolites 220 neuroendocrine mechanisms, activation of 222
microbiota–gut–brain (MGB) axis 53 neurohormonal signaling molecules 88
microbiota investigation tools 240–242 neurons
microbiota metabolism of polyphenols and health AgRP/NPY 220
effects 190–205 hypothalamic 221
microbiota modulation, role of functional food in pro-opiomelanocortin (POMC) 220
243–245 newborn intestinal colonization 76
microbiota of insects 105 next-generation DNA sequencing technologies 6
microbiota’s diversity 145 next-generation sequencing (NGS) 8, 241
microbiotic composition 73 NGS see next-generation sequencing (NGS)
microflora Niemann-Pick C1-Like 1 (NPC1L1) protein 131
commensal 213 nonalcoholic steatohepatitis 73
in digestive system 134 non-carbohydrate prebiotics 163–164
microfluidics systems and gut chip 102–103 polyphenols 163
micro-mass sequencing technique 55 short-chain fatty acids 164
micronutrients 226 nonculturable microorganisms 6
microorganisms 4, 214 nondigestible polysaccharides–fiber 82–84
evolutionary divergence of 5 non-flavonoids 186
neonatal’s ingestion of 76 nonparametric tests 31
nonculturable 6 nonsteroidal anti-inflammatory drugs
Microviridae 56 (NSAID) 46
to Caudovirales ratio 47 NovaSeq 6000 9
to Siphoviridae ratios NovaSeq Xp workflow 9
in gut 50 novel metabolic approaches, considerations on
milk kefir 143 limitations of 201–205
milk protein isolate (MPI) 90 novel prebiotic formulations 168–169
Index 263