Article
Article
Correspondence
[email protected] (J.Y.),
[email protected] (J.Q.),
[email protected] (W.Z.),
[email protected] (G.-H.L.)
In brief
A 50-year multi-tissue proteomic atlas
reveals transcriptome-proteome
decoupling, proteostasis decline with
amyloid accumulation, and
asynchronous aging clocks, identifying
circulating senoproteins contributing to
vascular and systemic aging.
Highlights
• A proteomic blueprint of human organs across 50-year aging
stages
ll
Article
Comprehensive human proteome profiles across
a 50-year lifespan reveal aging trajectories
and signatures
Yingjie Ding,1,6,15 Yuesheng Zuo,1,6,15 Bin Zhang,2,6,15 Yanling Fan,1,15 Gang Xu,4,5,15,16 Zhongyi Cheng,12,15
Shuai Ma,2,3,6,13,14,15 Shuaiqi Fang,2,6,15 Ao Tian,2,6 Dandan Gao,8 Xi Xu,4,5 Qiaoran Wang,1,6 Yaobin Jing,7
Mengmeng Jiang,2,3 Muzhao Xiong,1,6 Jiaming Li,1,6 Zichu Han,1,6 Shuhui Sun,9 Si Wang,10,14,16 Fuchu He,11,16
Jiayin Yang,4,5,* Jing Qu,2,3,6,9,13,14,* Weiqi Zhang,1,3,6,13,14,* and Guang-Hui Liu2,3,6,10,13,14,17,*
1China National Center for Bioinformation and Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
2State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
3Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
4Liver Transplant Center, Organ Transplant Center, West China Hospital of Sichuan University, Chengdu 610000, China
5Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital of Sichuan
324000, China
9Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing 100029, China
10Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Aging Translational
Medicine Center, Beijing Municipal Geriatric Medical Research Center, Beijing Key Laboratory of Environment and Aging, Xuanwu Hospital
Capital Medical University, Beijing 100053, China
11State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing
SUMMARY
Proteins are the cornerstone of life. However, the proteomic blueprint of aging across human tissues remains
uncharted. Here, we present a comprehensive proteomic and histological analysis of 516 samples from 13
human tissues spanning five decades. This dynamic atlas reveals widespread transcriptome-proteome de-
coupling and proteostasis decline, characterized by amyloid accumulation. Based on aging-associated pro-
tein changes, we developed tissue-specific proteomic age clocks and characterized organ-level aging trajec-
tories. Temporal analysis revealed an aging inflection around age 50, with blood vessels being a tissue that
ages early and is markedly susceptible to aging. We further defined a plasma proteomic signature of aging
that matches its tissue origins and identified candidate senoproteins, including GAS6, driving vascular and
systemic aging. Together, our findings lay the groundwork for a systems-level understanding of human aging
through the lens of proteins.
ll
Article
B C D E
F G H I
ll
Article
our understanding of the aging patterns within individual human gan chronometry and uncovering the nexus between multi-or-
organs remains fragmented, and the molecular mechanisms that gan aging, biological markers, chronic disease risks, and ther-
govern the complex interplay of multi-organ aging are largely un- apeutic targets.
charted territories.18–21
Aging trajectories differ across organs, yet these organs RESULTS
exhibit close interactions and act synergistically to promote
organismal aging.22,23 Our understanding of both the shared A half-century proteomic compendium of human organ
and organ-specific characteristics of aging, as well as the sys- aging
temic changes they collectively undergo, remains limited. For To elucidate the intricate tapestry of human aging, we embarked
the development of innovative interventions targeting aging, pre- on a journey, compiling a proteomic encyclopedia that spans a
cise metrics that capture both organ-specific aging and sys- spectrum of tissues from individuals aged 14 to 68, inclusive of
temic aging heterogeneity are essential.24–32 The absence of both genders (Figure 1A; Table S1). Our meticulously curated
biomarkers capable of reflecting the aging pace of various or- collection, reflecting functional diversity and aging relevance,
gans complicates the establishment of organ-specific biological encompasses tissues from seven bodily systems: cardiovascu-
age.33–37 Furthermore, our understanding of the spatiotemporal lar (heart and aorta), digestive (liver, pancreas, and intestine), im-
dynamics in multi-organ aging is inadequate, and there is a crit- mune (spleen and lymph node), endocrine (adrenal gland and
ical need to identify tissues sensitive to aging at different life white adipose), respiratory (lung), integumentary (skin), and
stages and transition points.22,38–42 musculoskeletal (muscle), along with blood samples.
Proteins, the orchestrators of cellular function, face unceasing Using state-of-the-art mass spectrometry,52,53,58 we per-
disruptions to homeostasis as they age.4,26,43–49 It is crucial to formed quantitative proteomic analyses on each sample,
detail the decline of proteostasis across the entire spectrum of accompanied by parallel transcriptomic assays. This rigorous
protein quality control.50 However, our understanding of proteo- approach yielded a comprehensive catalog of over 12,771
mic changes within aged human tissues remains strikingly distinct proteins, covering the human protein-coding genome
limited, impeding a deep understanding of the fundamental across all chromosomes (Figures 1B, 1C, and S1A; Table S1).
mechanisms of aging and their progression to disease. Utilizing Beyond high-abundance proteins essential for translation and
state-of-the-art proteomic methods to quantify thousands of metabolic functions, we identified a substantial number of low-
plasma proteins with precision provides a systematic approach abundance proteins across tissues, including transporters and
to unraveling molecular responses to aging.40,51–57 Yet, research hormone regulators (Figure 1D). We also profiled the character-
has mainly focused on plasma proteomics, often overlooking or- istics of detected proteins based on their subcellular localization,
gan-specific proteostasis disruptions, protein exchanges be- cellular component, and molecular function. The majority were
tween the vasculature and various organs, and the organ origins intracellular, with additional detection of membrane-bound and
of these blood-based proteomic shifts. secreted proteins. A large number localized to nuclear and
Our study is poised to construct a comprehensive multi-tis- organelle compartments, particularly mitochondria. Function-
sue proteomic atlas spanning 50 years of the entire human ag- ally, these proteins span diverse roles, including transcriptional
ing process, elucidating the mechanisms behind proteostasis and epigenetic regulation, as well as receptor-ligand signaling,
imbalance in aged organs and revealing both universal and tis- with notable heterogeneity across tissues (Figure 1E). Uniform
sue-specific aging patterns. This work aims to broaden our manifold approximation and projection (UMAP) unveiled highly
comprehension of the proteomic signatures of aging and to en- tissue-specific proteomic signatures, with discernible similarities
gineer organ aging clocks that pinpoint the onset tissues and between heart and skeletal muscle, as well as between spleen
pivotal time points. Additionally, we seek to explore the dy- and lymph node tissues (Figures S1B and S1C). We identified tis-
namic interplay between circulating and organ proteins during sue-enriched and tissue-enhanced proteins, as well as those
aging, with the goal of identifying systemic biomarkers for or- common across tissues, which are vital for basic housekeeping
Figure 1. A multi-tissue proteomic atlas outlines age- and tissue-associated protein features
(A) Schematic overview of the study. Samples spanning > 50 years of age were collected from 76 individuals across 13 tissues (n = 516). Deep proteomic,
transcriptomic, and histological profiling enabled the construction of a tissue-specific aging atlas, incorporating aging clocks and biomarkers.
(B) Left: number of proteins identified per tissue. Right: density distribution of protein size, and darker colors indicate higher counts.
(C) Chromosomal distribution of detected proteins across tissues, with red indicating higher density.
(D) Left: protein abundance heatmap across tissues. Right: Gene Ontology (GO) enrichment for high/low-abundance proteins. Blue to red: low to high abundance.
(E) Dot plot of detected proteins by subcellular localization, cellular component, and function across tissues. Dot size: relative proportion; color bar: protein count.
(F) Ring chart of proteins per tissue: dark gray (matched, protein with mRNA), light gray (unmatched, protein without mRNA).
(G) Density plots of mRNA-protein Spearman’s correlations per tissue.
(H) Violin plots of age-stratified Spearman’s correlations between mRNA and protein levels (only positively correlated proteins are shown).
(I) Left: negatively aging-related proteins in proteostasis. Right: positively correlated proteins (amyloids, immunoglobulins, and complement components). The
diagram on the far right illustrates the molecular interactions among these proteins that are supported by the literature. Bar plot shows gene set enrichment
analysis (GSEA) scores for selected pathways.
(J) Immunofluorescence (IF) staining of SAA1/2, IGLC, IGHA, and C4A in lymph, heart, and intestine (n = 22–27). Each point: one individual. Correlation by
Spearman’s test. Scale bars, 50 μm.
See also Figures S1 and S2.
ll
Article
A B
D E F
ll
Article
functions in biology. Tissue-enriched proteins, with their distinct and mitochondrial ribosomal proteins as well as aminoacyl-
expression profiles, underscore the specific physiological roles tRNA synthetases and peptidyl-tRNA hydrolase (Figure 1I).
of each tissue (Figures S1D–S1H; Table S1). Even within analo- This suggests a potential decline in translational capacity with
gous organs, our analysis precisely identified differentially ex- age. Post-translationally, proteins involved in folding, assembly,
pressed proteins (DEPs), as validated by western blotting for and transport, such as those in the chaperonin-containing
specific expressions, including SULT1B1, a liver xenobiotic- T-complex protein 1 subunit alpha (TCP1), Sec1/Munc18-family,
metabolizing enzyme59; PNLIPRP2, a pancreatic lipase that hy- endoplasmic reticulum membrane complex, and nuclear pore
drolyzes galactolipids60; aquaporin 4 (AQP4), a sarcolemma pro- complex, were found to decrease with aging (Figures 1I and
tein in muscle; and desmoglein 2 (DSG2), a desmosomal protein S2F). For example, histidyl-TRNA synthetase 1 (HARS1), a his-
in heart (Figure S1F).61,62 Additionally, we examined the impact tidyl-tRNA synthetase; chaperonin containing TCP1 subunit 5
of biological sex on protein expression across tissues and found (CCT5) and chaperonin containing TCP1 subunit 7 (CCT7), sub-
that only a few proteins exhibited sex-differentiated abundance, units of the ring-shaped chaperonin T-complex protein ring com-
such as the epigenetic regulator SGF29 in female hearts and the plex (TRiC); and proteasome subunit beta type-1 (PSMB1), a
matrix metalloprotease 3 (MMP3) in male white adipose tissue non-catalytic subunit of the 20S proteasome, were all reduced
(Figures S1I–S1K). at the protein level in aged tissues (Figure S2G). Consistent
with the decline in protein quality control, there is an accumula-
Aging disrupts mRNA-protein coupling and proteomic tion of amyloid proteins alongside immunoglobulin and comple-
homeostasis across tissues ment (Figure 1I). Proteomics data analysis and tissue immunoflu-
Protein homeostasis, a system encompassing the cellular mech- orescence analysis confirmed the accumulation of amyloid
anisms that govern protein synthesis, folding, modification, proteins, such as serum amyloid A1 (SAA1) and serum amyloid
translocation, and degradation, is a critical facet of cellular health A2 (SAA2), and the excessive activation of the immune pathway
(Figure S2A).43,44,63–65 To assess the transcriptional regulation of (such as immunoglobulins lambda constant 1 [IGLC], immuno-
proteomic patterns, we compared proteomic profiles with tran- globulin heavy constant alpha 1 [IGHA], and complement C4A)
scriptomic data. Over 95% of identified proteins showed a (Figures 1I and 1J). Considering that aberrant amyloid deposition
detectable transcriptional signal, yet the correlation between can trigger immunoglobulin activation, leading to the comple-
mRNA and protein levels was only moderately aligned (correla- ment pathway engagement,66–69 the amyloid-immunoglobulin-
tions ranged from 0.39 to 0.50) (Figures 1F and 1G). The positive complement axis may constitute a crucial component of the ag-
correlation diminishes with age, particularly pronounced in the ing tissue microenvironment.
spleen, muscle, and lymph nodes, underscoring an age-depen- We further observed increased variability in tissue-specific
dent decoupling between protein and transcription (Figures 1H protein expression in older individuals across organs, as indi-
and S2B–S2E; Table S2). Notably, these decoupled proteins cated by elevated standard deviation (SD) and coefficient of vari-
are pivotal for tissue functional maintenance, such as regulating ation (CV) in these proteins in aged samples (Figure S2H). In
B cell activation in the spleen (Figure S2D). These findings under- addition, we found a marked reduction in sex-biased proteomic
score the increasing disparity between mRNA and protein levels signatures in aged tissues (Figure S2I), suggesting a conver-
in aging organs, stressing the importance of understanding the gence of sex-specific expression patterns over time.
proteomic aspects of aging.
We next examined the key pathways across the entire protein Proteomic dynamics and biomarkers of aging in human
metabolism lifecycle, which may influence the protein quality organs
control capacity and subsequently cause mRNA-protein decou- To uncover the lifelong dynamics of the human proteome, we
pling during aging (Figure 1I). At the translational level, we focused on the proteins with differential abundance associated
observed a decrease in the abundance of key proteins involved with aging, identifying a substantial number of age-dependent
in protein synthesis across most tissues, including cytoplasmic that exhibited altered expression in at least one organ during
ll
Article
A B
C D
E F
ll
Article
human aging. Age-dependent DEPs were observed in various immune cell adhesion, and increases interleukin-6 (IL-6)
tissues, with some tissues showing pronounced sensitivity to ag- secretion (Figures S3G–S3K).
ing (Figures 2A, S3A, and S3B; Table S2). Through in-depth char-
acterization of the expression patterns of all DEPs across various Proteomic perturbation of biological pathways in aged
organs, we identified distinct modules for upregulated and human organs
downregulated DEPs. This analysis revealed organ-specific pro- To clarify proteomic shifts in aging organs, we established an ag-
tein signatures that reflect the dynamic changes associated with ing hallmark assessment system to track aging characteristics.
aging (Figures 2B, 2C, and S3C–S3F). Module 1 of upregulated This system includes markers for cell cycle arrest, genomic
DEPs prevalent across tissues and consistently rising with age instability, loss of heterochromatin, inflammation, and fibrosis
contains proteins involved in complement activation, immune (Figure 3A). In line with the proteomic data, histological staining
response, and amyloid fibril formation (Figure 2C), highlighting revealed elevated p21Cip1 expression, enhanced DNA damage
that these proteins form a ‘‘protein community’’ that synergisti- signals, reduced heterochromatin mark H3K9me3, increased
cally increases with aging across tissues. Other modules primar- pro-inflammatory factor IL-6, and alongside an elevation in tis-
ily consisted of tissue-specific DEPs that increased with age, sue fibrosis indicated by collagen IV in aged tissues, all of which
with corresponding pathways notably enriched in the aorta, are established biomarkers of aging (Figures 3B and S4A).
lung, heart, spleen, and lymph nodes, respectively (Figures 2C, Epigenetic instability is a key driver of aging, leading us to un-
S3C, and S3E). Regarding downregulated DEPs, we detected cover changes in epigenetic regulatory proteins associated
a steady decrease in proteins related to Golgi vesicle transport with aging, including those that regulate DNA methylation,
and protein maturation across distinct tissues. Other modules histone modification, chromatin remodeling, and RNA epige-
comprised tissue-specific DEPs that declined progressively netics (Figures 3C and S4B).9,26,70–80 Notably, YTHDC1 emerged
with age, with the downregulated pathways in various tissues as the most consistently downregulated epigenetic regulator
partially mirroring the aging-induced deterioration in tissue func- across tissues, and its reduced expression in aged liver and
tions (Figures 2C, S3D, and S3F). adrenal tissues was further validated by histological staining
Our analysis also revealed that while the majority of DEPs (Figure S4C). Among the aging hallmarks, we also observed
are tissue-specific, approximately 40% were found to be a widespread downregulation of mitochondrial proteins impli-
shared among at least two tissues (Figures 2D and 2E). cated in mitochondrial biogenesis, homeostasis, and respiration,
Notably, 31 proteins are uniformly downregulated across underscoring the importance of mitochondria in aging
over six tissues without upregulation in any organ. These (Figure 3D).
are termed ubiquitous downregulated proteins with aging Proteins often exert their functions within complexes, prompt-
(UDPAs), including pre-mRNA splicing tri-snRNP complex ing our quantitative dissection of the complexome to delineate
factor (PRPF4) involved in pre-mRNA splicing (Figures 2E aging-associated alterations. We analyzed a total of 816 protein
and 2F). Conversely, 29 proteins are consistently upregulated complexes and observed the downregulation of transcriptional
across more than six tissues without conflicting trends regulatory complexes (e.g., AR-NONO-SFPQ) and RNA splicing
(Figures 2E and 2F). These are designated ubiquitous upregu- complexes (e.g., spliceosome) across tissues (Figures 3E and
lated proteins with aging (UUPAs), associated with comple- S4D). By contrast, upregulated complexes were enriched in im-
ment activation, inflammatory response, and cell adhesion. mune and inflammatory responses (e.g., MAVS-TRADD) and
Among them, serum amyloid P-component (SAP, encoded extracellular matrix and cell adhesion (e.g., ITGA3-ITGB5) in
by APCS) is the most prominent UUPA, a canonical amyloid various tissues (Figure 3E). Gene set enrichment analysis
factor and a prime example of mRNA-protein decoupling (GSEA) corroborated these findings, revealing that upregulated
in the liver (Figures 2E, 2G, and S2E). Its elevation in multiple complexes were predominantly enriched in the inflammasome
aged tissues is confirmed by histological staining (Figure 2H). pathway, whereas downregulated complexes were linked to nu-
This protein’s upregulation with aging may promote vascular clear hormone receptors, nuclear transport, and spliceosomes
aging, damage, and inflammation, as treating human aortic (Figure 3F).
endothelial cells (hAECs) with recombinant SAP protein in- Finally, to elucidate the connection between tissue aging and
creases SA-β-Gal positivity, impairs tube formation, enhances chronic disease susceptibility, we constructed a protein-disease
Figure 3. Shared aging proteomic signatures converge on core pathways across tissues
(A) Ridge plots of age-correlated proteins from GSEA. Spearman’s correlation was applied. Normalized enrichment score (NES) indicated by color.
(B) IHC or IF staining of aging markers in tissues. p21Cip1, γH2AX, H3K9me3, IL-6, and collagen IV were quantified and correlated with age. Each point: one
individual. Sample sizes vary by marker and tissue. Spearman’s test was applied. Scale bars, 50 μm.
(C) Dot plot of GSEA score and enriched p values for epigenetic pathways. Dot size: − log10(p value); color bar: NES. *p < 0.05, **p < 0.01, ***p < 0.001. NES
indicated by color.
(D) Dot plot of GSEA score and enriched p values for mitochondrial pathways. Dot size: − log10(p value); color bar: NES. *p < 0.05, **p < 0.01, ***p < 0.001. NES
indicated by color.
(E) Heatmaps of upregulated (top) and downregulated (bottom) protein complexes across tissues. The top 15 shared complexes are highlighted. Blue to red: low
to high correlation coefficients.
(F) Left: GSEA of protein complex groups. Right: representative complex abundance with age correlation across tissues. Each point: one individual. Skin (n = 40),
aorta (n = 24). Spearman’s correlation was applied. NES indicated by color.
See also Figure S4.
ll
Article
A B
E F
G H I
ll
Article
interaction network by mapping aging-associated DEPs to Furthermore, sliding-window-based cumulative analysis re-
curated gene-disease annotations from the DisGeNET data- vealed that many tissues undergo substantial proteomic remod-
base.81 We revealed that the expression levels of 48 disease eling around age 50, particularly between 45 and 55 years
risk factors increased with age, encompassing a range of (Figures 4C, 4D, and S5I–S5K). Among all organs, the aorta ex-
chronic diseases, including cardiovascular conditions associ- hibited the most pronounced and continuous proteomic fluctua-
ated with APOE, CRP, ELN, and APOA1. Additionally, proteins tions across the lifespan. At age 30, the overall proportion of
like STAT3, TGFBR2, and MTOR are implicated in tissue fibrosis, DEPs remained low in most tissues, except for the aorta, spleen,
fatty liver, and hepatic tumors (Figure S4E).82–84 and adrenal gland, with the adrenal gland showing more signifi-
cant protein expression changes (Figures 4C and S5J). This sug-
Proteomic aging clocks for heterogeneous dynamics in gests that alterations in endocrine homeostasis may be one of
human organs the initiating events of systemic aging, consistent with our previ-
To quantitatively assess aging rate heterogeneity across human ous findings.24,89 At ages 45 and 55, the accumulation of DEPs
organs, we devised an innovative method for calculating the bio- intensified across organs, with the aorta showing the most prom-
logical age of each tissue (see STAR Methods) (Figure 4A). We inent changes, underscoring its exceptional sensitivity to aging.
established proteomic age clocks for 13 human tissues, expand- In parallel, the pancreas and spleen also exhibited sustained
ing and complementing previous clocks that solely depended on proteomic remodeling, likely reflecting aging-related deteriora-
plasma protein components.55,85,86 Employing the elastic net tion of metabolic and immune functions. Functionally, early alter-
regression algorithm, we identified proteins that precisely ations at age 30 were mainly associated with rRNA metabolic
predict each tissue’s chronological age, with Spearman’s corre- and translation, whereas transitions at ages 45 and 55 were en-
lation coefficients ranging from 0.74 to 0.95 (Figure 4B). We riched in RNA splicing and protein processing-related pathways
further dissected key proteins driving tissue-specific clocks (Figure S5K). These data collectively highlight the dynamic and
(Figures S5A–S5C; Table S3). Of particular interest, metallopro- heterogeneous nature of organ aging, with the vasculature
teinase inhibitor 3 (TIMP3), a regulator of metalloproteinase,87,88 emerging as one of the earliest and most affected systems.
was identified as a contributor to the tissue clocks across nine
distinct organs (Figures S5A–S5C). Our subsequent immuno- Tissue-specific profiling of secretory protein and inter-
staining confirmed that TIMP3 exhibits an age-associated in- tissue interactions
crease across various tissues (Figure S5D). Secretory proteins are key for extracellular functions and
This multi-time-point human organ protein navigator enabled mediate interactions among cells, organs, and systems.90–92
us to investigate critical transition points within the aging pro- We extracted secretory proteins from each organ to create an
cess. Through proteomic profiling across the human lifespan, age-related secretome dataset, comprising a total of 1,140 pro-
we pinpointed DEPs spanning the early, middle, and late teins. Our analysis included secretory proteins involved in immu-
stages of aging (Figures S5E–S5H). Despite their low preva- nity and inflammation (e.g., interleukins and chemokines), signal
lence, DEPs in early stages of aging associated with humoral transduction (e.g., neuropeptides and hormones), metabolism
immune responses and RNA splicing contribute to the aging and transport (e.g., apolipoproteins and enzymes), and struc-
DEPs across all age groups, with notable effects in the tural support (Figures S6A and S6B). We then categorized these
pancreas and aorta (Figures S5G and S5H). Middle-aged proteins into five modules based on their expression patterns,
DEPs constitute a substantial fraction of the lifespan-wide ag- which exhibited age-related changes (Figure S6C). The first
ing DEPs, underscoring the amplification of humoral immune module comprises secretory proteins that are universally
responses and complement activation across multiple tissues, increased across tissues, predominantly related to humoral im-
alongside the deregulation of RNA splicing in middle age mune responses and complement activation. The second mod-
(Figures S5G and S5H). ule includes proteins that are particularly upregulated in the small
ll
Article
A B C D E
F G H I J
K L M N
O P
Q S
ll
Article
intestine, mainly associated with immunoglobulin-mediated im- level. We identified 24 ligand-receptor pairs that mediate tissue
mune response and leukocyte-mediated immunity. The third communication via plasma and show age-related changes,
module consists of secretory proteins that are markedly upregu- mainly originating from the aorta and spleen. This highlights
lated in the aorta, regulating chemotaxis and blood coagulation. the key roles of vascular and immune tissues in orchestrating
The fourth module encompasses secretory proteins that are multi-organ aging (Figures 4H and 4I). Given that arteries age
specifically downregulated in the aorta, functioning in endothelial early and dramatically, as previously shown, these findings
cell migration and collagen fibril organization (Figure S6C). further support that arteries may be a key origin of systemic ag-
To decipher the intricacies of the senescence-associated ing (referred to as ‘‘senohub’’). They are not only highly sensitive
secretory phenotype (SASP), we have meticulously crafted tis- to aging-related changes but also serve as a major source of
sue-specific ‘‘SASP roadmaps’’ through an integrated analysis secretory senescence-promoting proteins (termed senopro-
of the aging-related secretome and the well-established teins), amplifying systemic aging signals.
SASP collection.42,93 Overall, we identified 579 distinct SASP Notably, growth arrest specific 6 (GAS6)—a senokine elevated
proteins, about half of them shared across at least two tissues in both plasma and its source tissue, the aorta—showed height-
(Figure S6D). CXCL12, a chemokine,94 accumulated in nine tis- ened interaction with the broadly expressed TAM receptors
sues (Figures 4E and S6D) and was validated by immunostaining (TYRO3, AXL, and MERTK) (Figures 4I and S6G). This interaction
in aged human tissues (Figure 4F). Henceforth, cytokines may activate pro-thrombotic signaling pathways, contributing
released by senescent tissues or cells are collectively termed to endothelial dysfunction and arteriopathic conditions
senokines. (Figures S6G and S6H).96–98
Secretory proteins are key mediators of inter-organ communi- To test this hypothesis, we first validated the age-related upre-
cation, yet our understanding of how these interactions change gulation of GAS6 in human vascular tissues and plasma using
with aging in human organs is limited. We profiled aging-associ- immunohistochemical staining and ELISA (Figures 5A and 5B).
ated secretory protein dynamics and mapped ligand-receptor in- We then treated hAECs and human vascular smooth muscle
teractions across tissues via CellPhoneDB (Figures 4G and cells (hVSMCs) with recombinant human GAS6 protein
S6E).95 Using this approach, we uncovered potential inter-organ (Figure 5C). GAS6 treatment in hAECs increased SA-β-Gal pos-
communication networks, with the aorta, immune tissues such itivity and p21Cip1 expression, impaired tube formation and
as the spleen and lymph nodes, and endocrine organs like the ad- migratory capacity, elevated IL-6 secretion, and enhanced im-
renal gland showing extensive interactions with other tissues. mune cell adhesion (Figures 5D–5I). Similarly, in hVSMCs, seno-
Notably, these inter-organ interactions intensify with age, primarily kine GAS6 accelerated cellular senescence and elevated inflam-
driven by receptor-ligand pairs involving the inflammatory matory cytokine levels (Figures 5J–5L).
chemokines CXCL12 and CXCL14 (Figure S6F; Table S4). By To evaluate the in vivo effects of circulating GAS6, we intrave-
contrast, aging-related reductions in inter-organ interactions nously administered recombinant mouse GAS6 to 8-month-old
were observed for ephrin A1 (EFNA1), ephrin B1 (EFNB1), and mice every 5 days for a total of four doses (Figure 5M). At the
ephrin B2 (EFNB2), members of the Ephrin family, which mediate physiological level, GAS6-treated mice exhibited impaired phys-
bidirectional signaling through Eph receptors and are involved in ical performance, including reduced grip strength, decreased
regulating cell migration, tissue boundary formation, and vascular wire-hang endurance, and compromised balance and coordina-
development (Figure S6F; Table S4). tion, as evidenced by increased foot slips and prolonged
Considering proteins mediate organ-organ interactions via the crossing times in balance beam tests (Figure 5N). Histological
bloodstream, we integrated plasma proteomics to elucidate analyses showed prominent markers of vascular aging, including
these long-range, plasma-mediated interactions at the protein elevated p21Cip1 expression, increased oxidative DNA damage
Figure 5. GAS6 promotes human endothelial senescence and mouse multi-organ aging
(A) IHC staining of vascular wall GAS6 in individuals of different ages (n = 23). Spearman’s correlation was applied. Scale bars, 50 μm.
(B) ELISA showing plasma GAS6 levels across ages (n = 62). Each point: one individual. Pearson’s correlation was applied.
(C) Schematic of GAS6 treatment in hAECs and hVSMCs.
(D–I) SA-β-Gal staining (D), western blot (E), tube formation (F), migration assay (G), IL-6 ELISA (H), and monocyte adhesion assay (I) in hAECs treated with vehicle
(n = 6 for D and F–I; n = 3 for E) or GAS6 (n = 6 for D and F–I; n = 3 for E). Scale bars, 100 μm.
(J) SA-β-Gal staining in hVSMCs treated with vehicle (n = 6) or GAS6 (n = 6). Scale bars, 200 μm.
(K) IF staining of H3K9me3 and LAP2 in hVSMCs treated with vehicle (n = 300 nuclei) or GAS6 (n = 300 nuclei). Scale bars, 20 μm.
(L) ELISA of IL-6 in hVSMCs treated with vehicle (n = 6) or GAS6 (n = 6).
(M) Schematic of in vivo GAS6 injection in 8-month-old mice (every 5 days, 4 doses), with behavioral and histological assays after the final dose. Image of
AlphaFold-predicted mouse GAS6 structure.
(N) Functional tests (grip strength, wire hang, beam-crossing time, and foot slips) in mice treated with vehicle (n = 7) or GAS6 (n = 10).
(O) Aortic staining for p21Cip1, 8-OHdG, MMTV/CD31, oil red O, S100A8, and vWF/CD31 (vehicle: n = 6–8, GAS6: n = 9–10). Scale bars, 20 and 5 μm (zoomed-in
image).
(P) Liver staining for p21Cip1, H3K9me3, H&E, and oil red O (vehicle: n = 7–8, GAS6: n = 10). Scale bars, 25 μm.
(Q) Spleen staining for H&E, p21Cip1, and IL-6 (vehicle: n = 7, GAS6: n = 10). Scale bars, 50 μm.
(R) Lymph staining for IL-6, S100A8, and γH2AX (vehicle: n = 7, GAS6: n = 6). Scale bars, 25 μm.
(S) Adrenal staining for IL-6 and S100A8 (vehicle: n = 7, GAS6: n = 9). Scale bars, 20 μm.
Data are shown as mean ± SEM. Two-tailed unpaired t test or Mann-Whitney test were applied (D–L and N–S).
ll
Article
(8-OHdG), and heightened expression of endogenous retrovirus algorithms, we developed a plasma protein proxy tissue aging
(MMTV) (Figure 5O).70 GAS6 treatment additionally promoted clock that achieves comparable age prediction accuracy to the
lipid deposition while upregulating expression of inflammatory tissue protein matrix-derived clock and differentiates aging
marker S100A8 and vascular damage marker von Willebrand across various tissues (see STAR Methods) (Figures S7D–S7H).
factor (vWF) within the aorta (Figure 5O).
Beyond vascular tissues, GAS6 promoted accelerated senes- Humoral senoproteins driving vascular and systemic
cence phenotypes in multiple target organs (Figures 5P–5S). In aging
the liver, GAS6 treatment increased p21Cip1 expression, reduced We next explored whether specific senoproteins accumulating in
H3K9me3 levels, stimulated immune infiltration, and lipid accu- plasma could drive vascular aging, given their direct interaction
mulation (Figure 5P). In the spleen, GAS6 disrupted the structural with the vascular wall, especially endothelial cells (Figures 7A
integrity of the white pulp and elevated p21Cip1 and IL-6 expres- and 7B). To test this hypothesis, we treated hAECs with recom-
sion (Figure 5Q). GAS6 administration elevated expression of in- binant forms of plasma proteins that increase with age
flammatory markers IL-6 and S100A8 in lymph nodes and adre- (Figure 7B). Seven proteins—GPNMB, COMP, HTRA1, SLPI,
nal glands, while increasing DNA damage marker γH2AX in IGFBP7, NEGR1, and NOTCH3—induced hallmark features of
lymph nodes (Figures 5R and 5S). Collectively, these findings vascular endothelial senescence. These features included
demonstrate that elevated GAS6 contributes to accelerated increased SA-β-Gal activity, elevated expression of p21Cip1,
vascular and systemic aging. and reduced levels of H3K9me3 and LAP2 (Figures 7C–7F,
S8A, and S8B). Elevated expression of pro-inflammatory cyto-
Circulating protein biomarkers predicting organ aging kines (IL-6 and IL-8) and endothelial dysfunction marker
Unveiling blood-based biomarkers for organ aging is essential for (VCAM1), impaired migration and tube formation, enhanced
non-invasive aging assessments. We integrated plasma and or- monocyte adhesion, and increased IL-6 secretion were also
gan proteomic data to identify proteins with concordant changes observed (Figures 7G–7J and S8C–S8E).
in both, thereby characterizing potential fluid biomarkers for organ Transcriptomic analysis further revealed that treatment with
aging (see STAR Methods). Employing this approach, we identi- these proteins activated canonical senescence-associated
fied a total of 211 plasma-tissue paired DEPs that vary with aging genes and upregulated pathways related to cellular senescence,
in plasma and show age-dependent changes in at least one tissue oxidative stress, and inflammation (Figure S8F). To determine
in the same direction as in plasma (Figures 6A, S7A, and S7B). The whether these effects extend beyond endothelial cells, we
highest number of plasma-tissue paired DEPs was observed in treated hVSMCs with GPNMB, HTRA1, and IGFBP7, which simi-
the aorta, adrenal glands, and spleen, highlighting their influence larly induced senescence phenotypes, including elevated SA-
on aging-related alterations in plasma protein composition β-Gal activity, reduced H3K9me3, compromised LAP2, and
(Figure S7A). Furthermore, we focused primarily on the upregu- increased IL-6 expression (Figures S8G–S8P). As an in vivo
lated DEPs. Approximately 40% of the plasma-tissue paired example to examine whether aging-associated plasma proteins
DEPs were uniquely present in a single tissue within our examined can drive vascular aging and subsequent systemic decline, we
range (Figures 6A, S7A, and S7B). Notably, several plasma-tissue tested GPNMB via intravenous injection in middle-aged mice.
paired DEPs were identified that correspond between plasma and GPNMB-treated mice exhibited reduced grip strength and
at least one tissue, and these proteins exhibit remarkable accu- impaired motor coordination and balance (Figure 7K). Histologi-
mulation in aging plasma. For example, GPNMB, a reported cal analysis revealed that GPNMB accelerated vascular aging,
senescence-associated antigen,99 has been confirmed by us marked by increased DNA oxidation, reactivated retrotranspo-
and other studies to accumulate in aging plasma.100,101 Addition- sons, cell cycle arrest in vascular wall cells, endothelial damage
ally, we identified a series of plasma-tissue paired aging bio- with upregulated vWF, and increased vascular inflammation and
markers, including COMP, CHI3L1, NOTCH3, ITLN1, EFEMP1, lipid accumulation (Figures 7L–7R). Together, these findings
HTRA1, and LTBP2, whose elevation in human plasma was further demonstrate that senoproteins secreted by aging tissues can
validated by ELISA (Figure 6B). We also confirmed the aging- amplify senescence-promoting signals, mediating vascular and
related upregulation of GPNMB, COMP, NOTCH3, HTRA1, and multi-organ degeneration.
LTBP2 proteins across multiple tissues through histological im-
munostaining, with the most significant increases observed in DISCUSSION
the aorta (Figure 6C).
In addition, we identified age-dependent downregulation of Human organ aging is a primary driver of numerous chronic dis-
plasma-tissue paired proteins, including HARS1, CCT5, and eases, yet the molecular mechanisms that govern its complexity
PSMB1 (Figure S7B). Notably, HARS1, a protein associated and heterogeneity across organs remain largely elusive.11,106–108
with neurodegenerative and autoimmune diseases,102,103 ex- Our study harnesses advanced quantitative proteomics to reveal
hibited the most widespread decline across tissues (Figure the protein underpinnings of organ aging, creating a pioneering
S7B). ELISA and immunohistochemistry further confirmed the atlas across human tissues. This work offers innovative and in-
age-related decrease of HARS1 in plasma and its reduction in depth perspectives for understanding, anticipating, and ad-
the aorta (Figures S2G and S7C). Drawing from our analyses, dressing aging with a protein-centric approach.
we posited that a plasma protein-based clock could act as a We introduce protein-based aging clocks and dynamic aging
surrogate for the organ clocks constructed from actual tissue trajectories for human organs, illuminating their biological age
protein matrices.24,104,105 Accordingly, using machine learning and disease risks. Notably, tissues generally exhibit an increase
ll
Article
in aging rate around 50 years, aligning with our established non- components. Integrating plasma and tissue data, we identified
invasive multidimensional aging clocks.24,27 Our study highlights age-accumulating humoral senoproteins—pro-inflammatory se-
the early and pronounced aging of the aorta, underscoring the nokines—that accelerate vascular and systemic decline. This re-
critical role of vascular senescence in initiating systemic aging veals that blood vessels, as the primary senohub, are not only
through its extensive interactions with other organs and blood extremely aging-sensing units and reservoirs for receiving
ll
Article
A B C
E F
G H
I J
K L M N
O P Q R
ll
Article
aging-related factors from peripheral tissues but are also signif- for future research. Demographic limitations in tissue sampling
icantly affected by organ aging itself. may also impact the generalizability of our findings. Despite
Our study also probed proteostasis decline, a key aging hall- these caveats, our research provides initial evidence that human
mark.45,109–113 We observed transcriptome-proteome decou- aging can be analyzed, quantified, and potentially modulated
pling, indicating reduced central dogma-based information through a protein-centric approach.
flow with age. We identified widespread amyloid protein aggre-
gation as a common aging trait across human organs and RESOURCE AVAILABILITY
confirmed SAP’s role in promoting vascular endothelial damage.
Lead contact
Amyloid-induced chronic inflammation is closely linked to the
Further information and requests for resources and reagents should be
onset of cardiovascular and cerebrovascular diseases.114–122 directed to Dr. Guang-Hui Liu ([email protected]).
Additionally, we have observed an increase in complement pro-
teins and immunoglobulins, which serve as a universal aging Materials availability
signature across tissues.26,42,123–125 Interestingly, SAP and This study did not generate new unique reagents.
immunoglobulin G (IgG) seem to utilize similar Fcγ receptor acti-
vation to mediate pro-inflammatory signaling.42,126,127,128 These Data and code availability
findings constitute an amyloid-centric inflammaging framework • The proteomics data of human tissues have been deposited at the iProX
underlying human organ aging and degeneration. database consortium. Bulk RNA sequencing (RNA-seq) data of human
tissues have been deposited at the Genome Sequence Archive (GSA) in
In translational medicine, our study offers new strategies for
the National Genomics Data Center, Beijing Institute of Genomics
protein-targeted geroprotective interventions.101,129 Identified (China National Center for Bioinformation) of the Chinese Academy of
aging-associated cell surface proteins may serve as seno-anti- Sciences. Accession numbers are listed in the key resources table.
gens for developing senolytic vaccines or CAR-T therapies to • This paper does not report original code.
clear senescent cells.99,129 Moreover, proteolysis targeting • Any additional information required to reanalyze the data reported in this
chimera (PROTAC)-mediated degradation or neutralizing anti- paper is available from the lead contact upon request.
bodies against soluble senoproteins could help block aging
drivers at the protein level. ACKNOWLEDGMENTS
In summary, the human aging protein navigator established in
this study not only reveals aging biomarkers but also identifies We thank the Aging Biomarker Consortium for their invaluable feedback and
organ-associated aging drivers, thereby enhancing our under- guidance throughout this study. We thank Tong Chen, Zhao Wang, Li Yi,
standing of the complex and heterogeneous biology of aging. and Zhenru Wu from West China Hospital for their assistance with human tis-
sue procurement and processing; Jingyi Jia, Jiayi Liu, and Feifei Liu for their
These insights may facilitate the development of targeted inter-
support in animal management; Ansheng Tan, Tingting Yan, Peng Yang,
ventions for aging and age-related diseases, paving the way to
Xiaoyan Sun, Beibei Liu, Ying Jing, and Kejie Zheng for their assistance with
improve the health of older adults. mouse tissue harvesting; Lei Bai, Jing Chen, Jing Lu, Qun Chu, Ying Yang,
Luyang Tian, Shangyi Qiao, Xiuping Li, and Xuewei Chen for their administra-
tive support; and Meng Wan and Ying Zhou from the Institute of Biophysics,
Limitations of the study Chinese Academy of Sciences, for their assistance with slide image acquisi-
Our study provides a comprehensive proteomic perspective on tion. We thank the Human Organ Physiopathology Emulation System
human aging, yet there is room for improvement. Though we (HOPE) for assisting in the development of human cell senescence models.
strived for representative sampling across age groups and We thank the Proteomic Navigator of the Human Body (π-HuB) Project for their
sexes, limitations in human tissue availability restricted sample support and guidance in proteomic analysis. The graphical elements in this
size and data completeness. This study represents a proof of study’s figures are sourced partially from BioRender.com, with some designed
by Yizhu Wang. This work was supported by the National Natural Science
concept, and further analysis of organ aging dynamics and aging
Foundation of China (82488301), the National Key R&D Program of China
clocks in larger cohorts is necessary to enhance its robustness. (2022YFA1103700), the National Natural Science Foundation of China
Notably, data from critical organs such as the brain, kidneys, and (82125011, 82361148131, 92168201, 82330044, 32341001, 82322025,
reproductive system were absent, highlighting important areas 32121001, 82192863, 82361148130, 8231101626, 82422031, 82271600,
ll
Article
82471586, 92468303, 82401822, and 32088101), the National Key R&D Pro- ○ Oil red O staining
gram of China (2020YFA0804000 and 2022YFA1103800), the NCD-Major Proj- ○ BODIPY staining
ect (2024ZD0530400), the STI2030-Major Projects (2021ZD0202400), the Bei- ○ RNA-seq library construction and data processing
jing Natural Science Foundation (JQ24044, Z240018, F251011, Z230011, ○ Quality control of proteomics data
5242024, and 5244046), the CAS Basic Research Program (YSBR-076 and ○ Identification of high and low abundance proteins
YSBR-012), the Strategic Priority Research Program of CAS (XDA0460403 ○ Classification of proteins based on chromosome location of protein-
and XDC0200000), XPLORER PRIZE (2021-1045), the Shenzhen Medical coding genes
Research Fund (C2406001 and D2401003), the Beijing Anzhen Hospital ○ Classification of proteins based on subcellular location, cellular
Research Fund (2024AZB3002), the CAS Informatization Plan (CAS- component, and molecular function
WX2022SDC-XK14), Beijing Medical Research Institute Pilot Project ○ Classification of proteins based on differential expression between
(JYY2023-13), the CAS Youth Interdisciplinary Team, the Key Lab of Alz- tissues
heimer’s Disease of Zhejiang Province (ZJAD-2024001), the Institute of ○ Identification of sex-biased proteins
Zoology Initiative Program (2023IOZ0202, 2024IOZ0103, and 2023IOZ0102), ○ Bulk RNA-seq data processing
the Youth Innovation Promotion Association of CAS (2022083), the Talent Pro- ○ Decoupling analysis of protein abundance and mRNA expression
grams of Capital Medical University (12500825 and 12300927), the Beijing Uni- ○ Translational noise analysis
versity Faculty Training Program (BPHR202203105), the CAST Young Elite ○ Gene set variation analysis (GSVA)
Scientists Program (2021QNRC001), the Hainan Provincial Natural Science ○ Identification of age-dependent differentially expressed proteins
Fellowship Initiative (2024PG0022), and the CAS International Partnership Pro- ○ Functional enrichment analysis
G.-H.L., W.Z., J.Q., and J.Y. designed the project and supervised all experi- ○ Age-group-specific DEPs analysis
ments. J.Y., Y.D., G.X., X.X., Q.W., M.J., M.X., and Z.H. performed human ○ Sliding window analysis of the senescence proteome
sample collection and clinical coordination. Y.D., Y.Z., B.Z., Y.F., Y.J., and ○ Analysis of secretory proteins
S.M. conducted proteomic profiling and data acquisition. Z.C. provided intel- ○ Module analysis of secretory proteins
lectual and technical support in proteomic mass spectrometry analysis. Y.Z., ○ Senescence-associated secretory phenotype (SASP) analysis
B.Z., and Y.D. carried out bioinformatic and statistical analysis. Y.Z., B.Z., Y. ○ Tissue-tissue interaction analysis
D., and J.L. developed multi-organ aging clock modeling. Y.D., Y.F., S.F., A. ○ Tissue-tissue interactions mediated by circulating proteins
T., and Z.H. completed tissue immunostaining and histopathology. Y.D., Y. ○ Disease enrichment analysis of tissue-tissue interacted protein pairs
F., S.F., and A.T. executed plasma protein functional validation, including ○ Plasma protein proxy aging clock analysis
in vitro and in vivo. Y.D., S.F., and D.G. performed plasma ELISA experiments. • QUANTIFICATION AND STATISTICAL ANALYSIS
S.M., S.S., S.W., and F.H. supervised the project. G.-H.L., W.Z., J.Q., Y.D., Y.
Z., B.Z., Y.F., S.M., S.F., and A.T. contributed to manuscript writing, review,
and editing. SUPPLEMENTAL INFORMATION
ll
Article
6. Kennedy, B.K., Berger, S.L., Brunet, A., Campisi, J., Cuervo, A.M., Epel, 21. Fu, X., Wang, S., Wu, Y., Sun, Y., Liu, W., Xi, X., Li, G.L., Liu, K., Yuan, W.,
E.S., Franceschi, C., Lithgow, G.J., Morimoto, R.I., Pessin, J.E., et al. et al.; Aging; Biomarker Consortium (2025). A biomarker framework for
(2014). Geroscience: linking aging to chronic disease. Cell 159, auditory system aging: the Aging Biomarker Consortium consensus
709–713. https://doi.org/10.1016/j.cell.2014.10.039. statement. Life Med. 4, lnaf011. https://doi.org/10.1093/lifemedi/
7. Prince, M.J., Wu, F., Guo, Y., Gutierrez Robledo, L.M., O’Donnell, M., lnaf011.
Sullivan, R., and Yusuf, S. (2015). The burden of disease in older people 22. Schaum, N., Lehallier, B., Hahn, O., Pálovics, R., Hosseinzadeh, S., Lee,
and implications for health policy and practice. Lancet 385, 549–562. S.E., Sit, R., Lee, D.P., Losada, P.M., Zardeneta, M.E., et al. (2020).
https://doi.org/10.1016/S0140-6736(14)61347-7. Ageing hallmarks exhibit organ-specific temporal signatures. Nature
8. Zhang, W., Che, Y., Tang, X., Chen, S., Song, M., Wang, L., Sun, A.J., 583, 596–602. https://doi.org/10.1038/s41586-020-2499-y.
Chen, H.Z., Xu, M., et al.; Aging; Biomarker Consortium (2023). A 23. Rando, T.A., and Wyss-Coray, T. (2021). Asynchronous, contagious and
biomarker framework for cardiac aging: the Aging Biomarker Consortium digital aging. Nat Aging 1, 29–35. https://doi.org/10.1038/s43587-020-
consensus statement. Life Med. 2, lnad035. https://doi.org/10.1093/life- 00015-1.
medi/lnad035.
24. Li, J., Xiong, M., Fu, X.H., Fan, Y., Dong, C., Sun, X., Zheng, F., Wang, S.
9. Kroemer, G., Maier, A.B., Cuervo, A.M., Gladyshev, V.N., Ferrucci, L., W., Liu, L., Xu, M., et al. (2023). Determining a multimodal aging clock in a
Gorbunova, V., Kennedy, B.K., Rando, T.A., Seluanov, A., Sierra, F., cohort of Chinese women. Med. 4, 825–848.e13. https://doi.org/10.
et al. (2025). From geroscience to precision geromedicine: Understand- 1016/j.medj.2023.06.010.
ing and managing aging. Cell 188, 2043–2062. https://doi.org/10.1016/j.
25. Zhao, R., Lu, H., Yuan, H., Chen, S., Xu, K., Zhang, T., Liu, Z., Jiang, Y.,
cell.2025.03.011.
Suo, C., and Chen, X. (2025). Plasma proteomics-based organ-specific
10. Guo, J., Huang, X., Dou, L., Yan, M., Shen, T., Tang, W., and Li, J. (2022). aging for all-cause mortality and cause-specific mortality: a prospective
Aging and aging-related diseases: from molecular mechanisms to inter- cohort study. GeroScience 47, 1411–1423. https://doi.org/10.1007/
ventions and treatments. Signal Transduct. Target. Ther. 7, 391. https:// s11357-024-01411-w.
doi.org/10.1038/s41392-022-01251-0.
26. Bao, H., Cao, J., Chen, M., Chen, M., Chen, W., Chen, X., Chen, Y., Chen,
11. Tian, Y.E., Cropley, V., Maier, A.B., Lautenschlager, N.T., Breakspear, Y., Chen, Y., et al.; Aging; Biomarker Consortium (2023). Biomarkers of
M., and Zalesky, A. (2023). Heterogeneous aging across multiple organ aging. Sci. China Life Sci. 66, 893–1066. https://doi.org/10.1007/
systems and prediction of chronic disease and mortality. Nat. Med. 29, s11427-023-2305-0.
1221–1231. https://doi.org/10.1038/s41591-023-02296-6.
27. Zheng, Z., Li, J., Liu, T., Fan, Y., Zhai, Q.C., Xiong, M., Wang, Q.R., Sun,
12. Suryadevara, V., Hudgins, A.D., Rajesh, A., Pappalardo, A., Karpova, A., X., Zheng, Q.W., Che, S., et al. (2024). DNA methylation clocks for esti-
Dey, A.K., Hertzel, A., Agudelo, A., Rocha, A., Soygur, B., et al. (2024). mating biological age in Chinese cohorts. Protein Cell 15, 575–593.
SenNet recommendations for detecting senescent cells in different tis- https://doi.org/10.1093/procel/pwae011.
sues. Nat. Rev. Mol. Cell Biol. 25, 1001–1023. https://doi.org/10.1038/
28. Sun, S., Jiang, M., Ma, S., Ren, J., and Liu, G.H. (2025). Exploring the het-
s41580-024-00738-8.
erogeneous targets of metabolic aging at single-cell resolution. Trends
13. Zheng, F., Lei, J., He, Z., Ning, T., Sun, S., Cai, Y., Zhao, Q., Ma, S., Endocrinol. Metab. 36, 133–146. https://doi.org/10.1016/j.tem.2024.
Zhang, W., Qu, J., et al. (2025). FOXO3-engineered human mesenchymal 07.009.
stem cells efficiently enhance post-ischemic stroke functional rehabilita-
tion. Protein Cell 16, 365–373. https://doi.org/10.1093/procel/pwaf004. 29. Ma, S., Chi, X., Cai, Y., Ji, Z., Wang, S., Ren, J., and Liu, G.H. (2023). De-
coding Aging Hallmarks at the Single-Cell Level. Annu. Rev. Biomed.
14. Yang, Y., Lu, X., Liu, N., Ma, S., Zhang, H., Zhang, Z., Yang, K., Jiang, M.,
Data Sci. 6, 129–152. https://doi.org/10.1146/annurev-biodatasci-
Zheng, Z., Qiao, Y., et al. (2024). Metformin decelerates aging clock in
020722-120642.
male monkeys. Cell 187, 6358–6378.e29. https://doi.org/10.1016/j.cell.
2024.08.021. 30. Liu, B., Qu, J., Zhang, W., Izpisua Belmonte, J.C., and Liu, G.H. (2022). A
stem cell aging framework, from mechanisms to interventions. Cell Rep.
15. Yan, H., Ren, J., and Liu, G.-H. (2023). Fecal microbiota transplantation:
41, 111451. https://doi.org/10.1016/j.celrep.2022.111451.
A new strategy to delay aging. hLife 1, 8–11. https://doi.org/10.1016/j.
hlife.2023.06.002. 31. Li, J., Zheng, Y., Yan, P., Song, M., Wang, S., Sun, L., Liu, Z., Ma, S., Iz-
pisua Belmonte, J.C., Chan, P., et al. (2021). A single-cell transcriptomic
16. Zhang, Z., Tian, X., Lu, J.Y., Boit, K., Ablaeva, J., Zakusilo, F.T., Emmrich,
atlas of primate pancreatic islet aging. Natl. Sci. Rev. 8, nwaa127. https://
S., Firsanov, D., Rydkina, E., Biashad, S.A., et al. (2023). Increased hya-
doi.org/10.1093/nsr/nwaa127.
luronan by naked mole-rat Has2 improves healthspan in mice. Nature
621, 196–205. https://doi.org/10.1038/s41586-023-06463-0. 32. Lei, J., Jiang, X., Li, W., Ren, J., Wang, D., Ji, Z., Wu, Z., Cheng, F., Cai,
Y., Yu, Z.R., et al. (2022). Exosomes from antler stem cells alleviate
17. Lei, J., Xin, Z., Liu, N., Ning, T., Jing, Y., Qiao, Y., He, Z., Jiang, M., Yang,
mesenchymal stem cell senescence and osteoarthritis. Protein Cell 13,
Y., Zhang, Z., et al. (2025). Senescence-resistant human mesenchymal
220–226. https://doi.org/10.1007/s13238-021-00860-9.
progenitor cells counter aging in primates. Cell. https://doi.org/10.
1016/j.cell.2025.05.021. 33. May, M. (2023). Biomarkers of aging remain elusive as researchers try to
slow the biological clock. Nat. Med. 29, 2673–2676. https://doi.org/10.
18. Suo, J., Gan, Y., Xie, Y., Xu, S., Wang, J., Chen, D., Chen, L., Deng, L.,
1038/s41591-023-02560-9.
Feng, S., et al.; Aging; Biomarker Consortium (2023). A framework of bio-
markers for skeletal aging: a consensus statement by the Aging 34. Moqri, M., Herzog, C., Poganik, J.R., Justice, J., Belsky, D.W., Higgins-
Biomarker Consortium. Life Med. 2, lnad045. https://doi.org/10.1093/ Chen, A., Moskalev, A., Fuellen, G., Cohen, A.A., et al.; Biomarkers; Ag-
lifemedi/lnad045. ing Consortium (2023). Biomarkers of aging for the identification and
evaluation of longevity interventions. Cell 186, 3758–3775. https://doi.
19. Zhang, L., Wang, Z., Zhang, Y., Ji, R., Li, Z., Zou, J., and Gao, B. (2024).
org/10.1016/j.cell.2023.08.003.
Regulatory cellular and molecular networks in the bone microenviron-
ment during aging. Life Med. 3, lnae019. https://doi.org/10.1093/life- 35. Ren, J., Song, M., Zhang, W., Cai, J.P., Cao, F., Cao, Z., Chan, P., Chen,
medi/lnae019. C., Chen, G., Chen, H.Z., et al. (2023). The Aging Biomarker Consortium
20. Lu, H., Jing, Y., Zhang, C., Ma, S., Zhang, W., Huang, D., Zhang, B., Zuo, represents a new era for aging research in China. Nat. Med. 29, 2162–
Y., Qin, Y., Liu, G.H., et al. (2024). Aging hallmarks of the primate ovary 2165. https://doi.org/10.1038/s41591-023-02444-y.
revealed by spatiotemporal transcriptomics. Protein Cell 15, 364–384. 36. Moqri, M., Herzog, C., Poganik, J.R., Ying, K., Justice, J.N., Belsky, D.W.,
https://doi.org/10.1093/procel/pwad063. Higgins-Chen, A.T., Chen, B.H., Cohen, A.A., Fuellen, G., et al. (2024).
ll
Article
Validation of biomarkers of aging. Nat. Med. 30, 360–372. https://doi.org/ coverage with narrow-window data-independent acquisition. Nat. Bio-
10.1038/s41591-023-02784-9. technol. 42, 1855–1866. https://doi.org/10.1038/s41587-023-02099-7.
37. Herzog, C.M.S., Goeminne, L.J.E., Poganik, J.R., Barzilai, N., Belsky, D. 53. Stewart, H.I., Grinfeld, D., Giannakopulos, A., Petzoldt, J., Shanley, T.,
W., Betts-LaCroix, J., Chen, B.H., Chen, M., Cohen, A.A., et al.; Bio- Garland, M., Denisov, E., Peterson, A.C., Damoc, E., Zeller, M., et al.
markers; Aging Consortium (2024). Challenges and recommendations (2023). Parallelized Acquisition of Orbitrap and Astral Analyzers Enables
for the translation of biomarkers of aging. Nat Aging 4, 1372–1383. High-Throughput Quantitative Analysis. Anal. Chem. 95, 15656–15664.
https://doi.org/10.1038/s43587-024-00683-3. https://doi.org/10.1021/acs.analchem.3c02856.
38. Shen, X., Wang, C., Zhou, X., Zhou, W., Hornburg, D., Wu, S., and 54. Eldjarn, G.H., Ferkingstad, E., Lund, S.H., Helgason, H., Magnusson, O.
Snyder, M.P. (2024). Nonlinear dynamics of multi-omics profiles during T., Gunnarsdottir, K., Olafsdottir, T.A., Halldorsson, B.V., Olason, P.I.,
human aging. Nat Aging 4, 1619–1634. https://doi.org/10.1038/ Zink, F., et al. (2023). Large-scale plasma proteomics comparisons
s43587-024-00692-2. through genetics and disease associations. Nature 622, 348–358.
39. Wolf, J., Rasmussen, D.K., Sun, Y.J., Vu, J.T., Wang, E., Espinosa, C., Bi- https://doi.org/10.1038/s41586-023-06563-x.
gini, F., Chang, R.T., Montague, A.A., Tang, P.H., et al. (2023). Liquid-bi- 55. Oh, H.S.H., Rutledge, J., Nachun, D., Pálovics, R., Abiose, O., Moran-
opsy proteomics combined with AI identifies cellular drivers of eye aging Losada, P., Channappa, D., Urey, D.Y., Kim, K., Sung, Y.J., et al.
and disease in vivo. Cell 186, 4868–4884.e12. https://doi.org/10.1016/j. (2023). Organ aging signatures in the plasma proteome track health
cell.2023.09.012. and disease. Nature 624, 164–172. https://doi.org/10.1038/s41586-
40. Shen, Y., Timsina, J., Heo, G., Beric, A., Ali, M., Wang, C., Yang, C., 023-06802-1.
Wang, Y., Western, D., Liu, M., et al. (2024). CSF proteomics identifies 56. Ogrodnik, M., Carlos Acosta, J., Adams, P.D., d’Adda di Fagagna, F.,
early changes in autosomal dominant Alzheimer’s disease. Cell 187, Baker, D.J., Bishop, C.L., Chandra, T., Collado, M., Gil, J., Gorgoulis,
6309–6326.e15. https://doi.org/10.1016/j.cell.2024.08.049. V., et al. (2024). Guidelines for minimal information on cellular senes-
41. Li, L., Xu, Y., Yan, L., Li, X., Li, F., Liu, Z., Zhang, C., Lou, Y., Gao, D., cence experimentation in vivo. Cell 187, 4150–4175. https://doi.org/10.
Cheng, X., et al. (2023). Dynamic network biomarker factors orchestrate 1016/j.cell.2024.05.059.
cell-fate determination at tipping points during hESC differentiation. 57. Wilhelm, M., Schlegl, J., Hahne, H., Gholami, A.M., Lieberenz, M., Savit-
Innovation (Camb.) 4, 100364. https://doi.org/10.1016/j.xinn.2022. ski, M.M., Ziegler, E., Butzmann, L., Gessulat, S., Marx, H., et al. (2014).
100364. Mass-spectrometry-based draft of the human proteome. Nature 509,
42. Ma, S., Ji, Z., Zhang, B., Geng, L., Cai, Y., Nie, C., Li, J., Zuo, Y., Sun, Y., 582–587. https://doi.org/10.1038/nature13319.
Xu, G., et al. (2024). Spatial transcriptomic landscape unveils immuno- 58. Niu, L., Thiele, M., Geyer, P.E., Rasmussen, D.N., Webel, H.E., Santos,
globin-associated senescence as a hallmark of aging. Cell 187, 7025– A., Gupta, R., Meier, F., Strauss, M., Kjaergaard, M., et al. (2022). Nonin-
7044.e34. https://doi.org/10.1016/j.cell.2024.10.019. vasive proteomic biomarkers for alcohol-related liver disease. Nat. Med.
43. Kaushik, S., and Cuervo, A.M. (2015). Proteostasis and aging. Nat. Med. 28, 1277–1287. https://doi.org/10.1038/s41591-022-01850-y.
21, 1406–1415. https://doi.org/10.1038/nm.4001. 59. Choughule, K.V., Locuson, C.W., and Coughtrie, M.W.H. (2015). Charac-
44. Hipp, M.S., Kasturi, P., and Hartl, F.U. (2019). The proteostasis network terization of bovine phenol sulfotransferases: evidence of a major role for
and its decline in ageing. Nat. Rev. Mol. Cell Biol. 20, 421–435. https:// SULT1B1 in the liver. Xenobiotica 45, 495–502. https://doi.org/10.3109/
doi.org/10.1038/s41580-019-0101-y. 00498254.2014.997325.
45. López-Otı́n, C., Blasco, M.A., Partridge, L., Serrano, M., and Kroemer, G. 60. Zhu, G., Fang, Q., Zhu, F., Huang, D., and Yang, C. (2021). Structure and
(2023). Hallmarks of aging: An expanding universe. Cell 186, 243–278. Function of Pancreatic Lipase-Related Protein 2 and Its Relationship
https://doi.org/10.1016/j.cell.2022.11.001. With Pathological States. Front. Genet. 12, 693538. https://doi.org/10.
3389/fgene.2021.693538.
46. Aebersold, R., and Mann, M. (2016). Mass-spectrometric exploration of
proteome structure and function. Nature 537, 347–355. https://doi.org/ 61. Frigeri, A., Nicchia, G.P., Balena, R., Nico, B., and Svelto, M. (2004).
10.1038/nature19949. Aquaporins in skeletal muscle: reassessment of the functional role of
47. Keele, G.R., Zhang, J.G., Szpyt, J., Korstanje, R., Gygi, S.P., Churchill, G. aquaporin-4. FASEB J. 18, 905–907. https://doi.org/10.1096/fj.03-
A., and Schweppe, D.K. (2023). Global and tissue-specific aging effects 0987fje.
on murine proteomes. Cell Rep. 42, 112715. https://doi.org/10.1016/j. 62. Zhang, B., Wu, Y., Yang, X., Xiang, Y., and Yang, B. (2023). Molecular
celrep.2023.112715. insight into arrhythmogenic cardiomyopathy caused by DSG2 mutations.
48. Kim, M.S., Pinto, S.M., Getnet, D., Nirujogi, R.S., Manda, S.S., Chaer- Biomed. Pharmacother. 167, 115448. https://doi.org/10.1016/j.biopha.
kady, R., Madugundu, A.K., Kelkar, D.S., Isserlin, R., Jain, S., et al. 2023.115448.
(2014). A draft map of the human proteome. Nature 509, 575–581. 63. Wei, Y.-N., Hu, H.-Y., Xie, G.-C., Fu, N., Ning, Z.-B., Zeng, R., and Khai-
https://doi.org/10.1038/nature13302. tovich, P. (2015). Transcript and protein expression decoupling reveals
49. He, F., Aebersold, R., Baker, M.S., Bian, X., Bo, X., Chan, D.W., Chang, RNA binding proteins and miRNAs as potential modulators of human ag-
C., Chen, L., Chen, X., Chen, Y.J., et al. (2024). π-HuB: the proteomic ing. Genome Biol. 16, 41. https://doi.org/10.1186/s13059-015-0608-2.
navigator of the human body. Nature 636, 322–331. https://doi.org/10. 64. Cai, N., Li, M., Qu, J., Liu, G.H., and Izpisua Belmonte, J.C. (2012). Post-
1038/s41586-024-08280-5. translational modulation of pluripotency. J. Mol. Cell Biol. 4, 262–265.
50. Yan, P., Ren, J., Zhang, W., Qu, J., and Liu, G.H. (2020). Protein quality https://doi.org/10.1093/jmcb/mjs031.
control of cell stemness. Cell Regen. 9, 22. https://doi.org/10.1186/ 65. Cai, N., Li, M., Qu, J., Liu, G.H., and Izpisua Belmonte, J.C. (2012). Post-
s13619-020-00064-2. translational modulation of pluripotency. J. Mol. Cell Biol. 4, 262–265.
51. Sun, B.B., Chiou, J., Traylor, M., Benner, C., Hsu, Y.H., Richardson, T.G., https://doi.org/10.1093/jmcb/mjs031.
Surendran, P., Mahajan, A., Robins, C., Vasquez-Grinnell, S.G., et al. 66. Rahman, M.M., and Lendel, C. (2021). Extracellular protein components
(2023). Plasma proteomic associations with genetics and health in the of amyloid plaques and their roles in Alzheimer’s disease pathology. Mol.
UK Biobank. Nature 622, 329–338. https://doi.org/10.1038/s41586- Neurodegener. 16, 59. https://doi.org/10.1186/s13024-021-00465-0.
023-06592-6. 67. Schartz, N.D., and Tenner, A.J. (2020). The good, the bad, and the oppor-
52. Guzman, U.H., Martinez-Val, A., Ye, Z., Damoc, E., Arrey, T.N., Pash- tunities of the complement system in neurodegenerative disease.
kova, A., Renuse, S., Denisov, E., Petzoldt, J., Peterson, A.C., et al. J. Neuroinflammation 17, 354. https://doi.org/10.1186/s12974-020-
(2024). Ultra-fast label-free quantification and comprehensive proteome 02024-8.
ll
Article
68. Reichwald, J., Danner, S., Wiederhold, K.H., and Staufenbiel, M. (2009). framework for liver aging: the Aging Biomarker Consortium consensus
Expression of complement system components during aging and amy- statement. Life Med. 3, lnae004. https://doi.org/10.1093/lifemedi/
loid deposition in APP transgenic mice. J. Neuroinflammation 6, 35. lnae004.
https://doi.org/10.1186/1742-2094-6-35. 85. Argentieri, M.A., Xiao, S., Bennett, D., Winchester, L., Nevado-Holgado,
69. Doni, A., Parente, R., Laface, I., Magrini, E., Cunha, C., Colombo, F.S., A.J., Ghose, U., Albukhari, A., Yao, P., Mazidi, M., Lv, J., et al. (2024). Pro-
Lacerda, J.F., Campos, A., Jr., Mapelli, S.N., Petroni, F., et al. (2021). teomic aging clock predicts mortality and risk of common age-related
Serum amyloid P component is an essential element of resistance diseases in diverse populations. Nat. Med. 30, 2450–2460. https://doi.
against Aspergillus fumigatus. Nat. Commun. 12, 3739. https://doi.org/ org/10.1038/s41591-024-03164-7.
10.1038/s41467-021-24021-y. 86. Kuo, C.L., Chen, Z., Liu, P., Pilling, L.C., Atkins, J.L., Fortinsky, R.H., Ku-
70. Liu, X., Liu, Z., Wu, Z., Ren, J., Fan, Y., Sun, L., Cao, G., Niu, Y., Zhang, B., chel, G.A., and Diniz, B.S. (2024). Proteomic aging clock (PAC) predicts
Ji, Q., et al. (2023). Resurrection of endogenous retroviruses during aging age-related outcomes in middle-aged and older adults. Aging Cell 23,
reinforces senescence. Cell 186, 287–304.e26. https://doi.org/10.1016/j. e14195. https://doi.org/10.1111/acel.14195.
cell.2022.12.017. 87. Lee, W.T., Wu, P.Y., Cheng, Y.M., and Huang, Y.F. (2024). Tissue Inhib-
71. Li, M.H., Jiang, X., Jing, Y., Yan, K., Bi, S.J., Wang, S., Ma, S., Liu, G.H., itor of Metalloproteinase 3: Unravelling Its Biological Function and Signif-
Zhang, W., Sun, S., et al. (2025). CRISPR-based screening pinpoints icance in Oncology. Int. J. Mol. Sci. 25, 3191. https://doi.org/10.3390/
H2AZ1 as a driver of senescence in human mesenchymal stem cells. ijms25063191.
Protein Cell 16, 296–305. https://doi.org/10.1093/procel/pwae035. 88. Qi, J.H., and Anand-Apte, B. (2015). Tissue inhibitor of
72. Zhang, W., Qu, J., Liu, G.H., and Belmonte, J.C.I. (2020). The ageing epi- metalloproteinase-3 (TIMP3) promotes endothelial apoptosis via a cas-
genome and its rejuvenation. Nat. Rev. Mol. Cell Biol. 21, 137–150. pase-independent mechanism. Apoptosis 20, 523–534. https://doi.org/
https://doi.org/10.1038/s41580-019-0204-5. 10.1007/s10495-014-1076-y.
73. Wu, Z., Zhang, W., Qu, J., and Liu, G.H. (2024). Emerging epigenetic in- 89. Wang, Q., Wang, X., Liu, B., Ma, S., Zhang, F., Sun, S., Jing, Y., Fan, Y.,
sights into aging mechanisms and interventions. Trends Pharmacol. Sci. Ding, Y., Xiong, M., et al. (2024). Aging induces region-specific dysregu-
45, 157–172. https://doi.org/10.1016/j.tips.2023.12.002. lation of hormone synthesis in the primate adrenal gland. Nat Aging 4,
74. Ren, R., Deng, L., Xue, Y., Suzuki, K., Zhang, W., Yu, Y., Wu, J., Sun, L., 396–413. https://doi.org/10.1038/s43587-024-00588-1.
Gong, X., Luan, H., et al. (2017). Visualization of aging-associated chro- 90. Jiao, H., Li, X., Li, Y., Guo, Y., Hu, X., Sho, T., Luo, Y., Wang, J., Cao, H.,
matin alterations with an engineered TALE system. Cell Res. 27, Du, W., et al. (2024). Localized, highly efficient secretion of signaling pro-
483–504. https://doi.org/10.1038/cr.2017.18. teins by migrasomes. Cell Res. 34, 572–585. https://doi.org/10.1038/
75. Liang, C., Ke, Q., Liu, Z., Ren, J., Zhang, W., Hu, J., Wang, Z., Chen, H., s41422-024-00992-7.
Xia, K., Lai, X., et al. (2022). BMAL1 moonlighting as a gatekeeper for 91. Chen, J., Fang, M., Li, Y., Ding, H., Zhang, X., Jiang, X., Zhang, J., Zhang,
LINE1 repression and cellular senescence in primates. Nucleic Acids C., Lu, Z., and Luo, M. (2024). Cell surface protein–protein interaction
Res. 50, 3323–3347. https://doi.org/10.1093/nar/gkac146. profiling for biological network analysis and novel target discovery. Life
76. Wu, Z., Qu, J., and Liu, G.-H. (2024). Roles of chromatin and genome Med. 3, lnae031. https://doi.org/10.1093/lifemedi/lnae031.
instability in cellular senescence and their relevance to ageing and 92. Feizi, A., Gatto, F., Uhlen, M., and Nielsen, J. (2017). Human protein
related diseases. Nat. Rev. Mol. Cell Biol. 25, 979–1000. https://doi. secretory pathway genes are expressed in a tissue-specific pattern to
org/10.1038/s41580-024-00775-3. match processing demands of the secretome. npj Syst. Biol. Appl. 3,
77. Fan, Y., Zheng, Y., Zhang, Y., Xu, G., Liu, C., Hu, J., Ji, Q., Zhang, S., 22. https://doi.org/10.1038/s41540-017-0021-4.
Fang, S., Lei, J., et al. (2025). ARID5A orchestrates cardiac aging and 93. De Cecco, M., Ito, T., Petrashen, A.P., Elias, A.E., Skvir, N.J., Criscione,
inflammation through MAVS mRNA stabilization. Nat CardioVasc Res S.W., Caligiana, A., Brocculi, G., Adney, E.M., Boeke, J.D., et al. (2019).
4, 602–623. https://doi.org/10.1038/s44161-025-00635-z. L1 drives IFN in senescent cells and promotes age-associated inflamma-
78. Wang, C., Jiang, X., Li, H.Y., Hu, J., Ji, Q., Wang, Q., Liu, X., Huang, D., tion. Nature 566, 73–78. https://doi.org/10.1038/s41586-018-0784-9.
Yan, K., Zhao, L., et al. (2025). RIG-I-driven CDKN1A stabilization rein- 94. Biswas, L., Chen, J., De Angelis, J., Singh, A., Owen-Woods, C., Ding, Z.,
forces cellular senescence. Sci. China Life Sci. 68, 1646–1661. https:// Pujol, J.M., Kumar, N., Zeng, F., Ramasamy, S.K., et al. (2023).
doi.org/10.1007/s11427-024-2844-8. Lymphatic vessels in bone support regeneration after injury. Cell 186,
79. Wu, Z., Qu, J., Zhang, W., and Liu, G.H. (2024). Stress, epigenetics, and 382–397.e24. https://doi.org/10.1016/j.cell.2022.12.031.
aging: Unraveling the intricate crosstalk. Mol. Cell 84, 34–54. https://doi. 95. Garcia-Alonso, L., Lorenzi, V., Mazzeo, C.I., Alves-Lopes, J.P., Roberts,
org/10.1016/j.molcel.2023.10.006. K., Sancho-Serra, C., Engelbert, J., Marec �ková, M., Gruhn, W.H., Bot-
80. Zhao, H., Ji, Q., Wu, Z., Wang, S., Ren, J., Yan, K., Wang, Z., Hu, J., Chu, ting, R.A., et al. (2022). Single-cell roadmap of human gonadal develop-
Q., Hu, H., et al. (2022). Destabilizing heterochromatin by APOE mediates ment. Nature 607, 540–547. https://doi.org/10.1038/s41586-022-
senescence. Nat. Aging 2, 303–316. https://doi.org/10.1038/s43587- 04918-4.
022-00186-z. 96. Wu, G., Ma, Z., Cheng, Y., Hu, W., Deng, C., Jiang, S., Li, T., Chen, F., and
81. Piñero, J., Ramı́rez-Anguita, J.M., Saüch-Pitarch, J., Ronzano, F., Cen- Yang, Y. (2018). Targeting Gas6/TAM in cancer cells and tumor microen-
teno, E., Sanz, F., and Furlong, L.I. (2020). The DisGeNET knowledge vironment. Mol. Cancer 17, 20. https://doi.org/10.1186/s12943-018-
platform for disease genomics: 2019 update. Nucleic Acids Res. 48, 0769-1.
D845–D855. https://doi.org/10.1093/nar/gkz1021. 97. Happonen, K.E., and Dahlbäck, B. (2016). Gas6 fueling tumor-mediated
82. Ye, Y., Yang, K., Liu, H., Yu, Y., Song, M., Huang, D., Lei, J., Zhang, Y., thrombosis. Blood 127, 672–673. https://doi.org/10.1182/blood-2015-
Liu, Z., Chu, Q., et al. (2023). SIRT2 counteracts primate cardiac aging 12-683474.
via deacetylation of STAT3 that silences CDKN2B. Nat Aging 3, 1269– 98. Borgel, D. (2008). Gas6 inflames cell interactions. Blood 111, 3915.
1287. https://doi.org/10.1038/s43587-023-00486-y. https://doi.org/10.1182/blood-2008-01-133314.
83. Chen, C., Liu, J., and Yu, W. (2024). Unlocking heart anti-aging potential: 99. Suda, M., Shimizu, I., Katsuumi, G., Yoshida, Y., Hayashi, Y., Ikegami, R.,
the SIRT2-STAT3-CDKN2B pathway as a bridge between fiction and re- Matsumoto, N., Yoshida, Y., Mikawa, R., Katayama, A., et al. (2021). Se-
ality. Life Med. 3, lnae020. https://doi.org/10.1093/lifemedi/lnae020. nolytic vaccination improves normal and pathological age-related phe-
84. Jiang, M., Zheng, Z., Wang, X., Chen, Y., Qu, J., Ding, Q., Zhang, W., Liu, notypes and increases lifespan in progeroid mice. Nat Aging 1, 1117–
Y.S., Yang, J., et al.; Aging; Biomarker Consortium (2024). A biomarker 1126. https://doi.org/10.1038/s43587-021-00151-2.
ll
Article
100. Liu, Y.Y., Pang, J., Zhang, C., Zeng, L.T., Wang, Y., Wang, S.B., Fan, G. 117. Gillmore, J.D., and Hawkins, P.N. (2013). Pathophysiology and treatment
Q., Zhang, L.Q., Shen, T., Li, X.F., et al. (2024). Biofluid GPNMB/osteoac- of systemic amyloidosis. Nat. Rev. Nephrol. 9, 574–586. https://doi.org/
tivin as a potential biomarker of ageing: A cross-sectional study. Heliyon 10.1038/nrneph.2013.171.
10, e36574. https://doi.org/10.1016/j.heliyon.2024.e36574. 118. Skovronsky, D.M., Lee, V.M.Y., and Trojanowski, J.Q. (2006). Neurode-
101. Sun, S., Li, J., Wang, S., Li, J., Ren, J., Bao, Z., Sun, L., Ma, X., Zheng, F., generative diseases: new concepts of pathogenesis and their therapeu-
Ma, S., et al. (2023). CHIT1-positive microglia drive motor neuron ageing tic implications. Annu. Rev. Pathol. 1, 151–170. https://doi.org/10.1146/
in the primate spinal cord. Nature 624, 611–620. https://doi.org/10.1038/ annurev.pathol.1.110304.100113.
s41586-023-06783-1. 119. Iadanza, M.G., Jackson, M.P., Hewitt, E.W., Ranson, N.A., and Radford,
102. Adams, R.A., Fernandes-Cerqueira, C., Notarnicola, A., Mertsching, E., S.E. (2018). A new era for understanding amyloid structures and disease.
Xu, Z., Lo, W.S., Ogilvie, K., Chiang, K.P., Ampudia, J., Rosengren, S., Nat. Rev. Mol. Cell Biol. 19, 755–773. https://doi.org/10.1038/s41580-
et al. (2021). Serum-circulating His-tRNA synthetase inhibits organ-tar- 018-0060-8.
geted immune responses. Cell. Mol. Immunol. 18, 1463–1475. https:// 120. Gao, Y., Hong, Y., Huang, L., Zheng, S., Zhang, H., Wang, S., Yao, Y.,
doi.org/10.1038/s41423-019-0331-0. Zhao, Y., Zhu, L., Xu, Q., et al. (2023). β2-microglobulin functions as an
endogenous NMDAR antagonist to impair synaptic function. Cell 186,
103. Galatolo, D., Kuo, M.E., Mullen, P., Meyer-Schuman, R., Doccini, S., Bat-
1026–1038.e20. https://doi.org/10.1016/j.cell.2023.01.021.
tini, R., Lieto, M., Tessa, A., Filla, A., Francklyn, C., et al. (2020). Bi-allelic
mutations in HARS1 severely impair histidyl-tRNA synthetase expression 121. Langworth-Green, C., Patel, S., Jaunmuktane, Z., Jabbari, E., Morris, H.,
and enzymatic activity causing a novel multisystem ataxic syndrome. Thom, M., Lees, A., Hardy, J., Zandi, M., and Duff, K. (2023). Chronic ef-
Hum. Mutat. 41, 1232–1237. https://doi.org/10.1002/humu.24024. fects of inflammation on tauopathies. Lancet Neurol. 22, 430–442.
https://doi.org/10.1016/S1474-4422(23)00038-8.
104. Ozcelik, A., Rufo, J., Guo, F., Gu, Y., Li, P., Lata, J., and Huang, T.J.
(2018). Acoustic tweezers for the life sciences. Nat. Methods 15, 1021– 122. Richards, D.B., Cookson, L.M., Berges, A.C., Barton, S.V., Lane, T., Rit-
1028. https://doi.org/10.1038/s41592-018-0222-9. ter, J.M., Fontana, M., Moon, J.C., Pinzani, M., Gillmore, J.D., et al.
(2015). Therapeutic Clearance of Amyloid by Antibodies to Serum Amy-
105. Rutledge, J., Oh, H., and Wyss-Coray, T. (2022). Measuring biological
loid P Component. N. Engl. J. Med. 373, 1106–1114. https://doi.org/10.
age using omics data. Nat. Rev. Genet. 23, 715–727. https://doi.org/
1056/NEJMoa1504942.
10.1038/s41576-022-00511-7.
123. Ma, S., Sun, S., Geng, L., Song, M., Wang, W., Ye, Y., Ji, Q., Zou, Z.,
106. Moqri, M., and Snyder, M. (2023). Organ-specific aging and the risk of Wang, S., He, X., et al. (2020). Caloric Restriction Reprograms the
chronic diseases. Nat. Med. 29, 1068–1069. https://doi.org/10.1038/ Single-Cell Transcriptional Landscape of Rattus Norvegicus Aging. Cell
s41591-023-02338-z. 180, 984–1001.e22. https://doi.org/10.1016/j.cell.2020.02.008.
107. Furman, D., Campisi, J., Verdin, E., Carrera-Bastos, P., Targ, S., France- 124. Yu, L., Wan, Q., Liu, Q., Fan, Y., Zhou, Q., Skowronski, A.A., Wang, S.,
schi, C., Ferrucci, L., Gilroy, D.W., Fasano, A., Miller, G.W., et al. (2019). Shao, Z., Liao, C.Y., Ding, L., et al. (2024). IgG is an aging factor that
Chronic inflammation in the etiology of disease across the life span. Nat. drives adipose tissue fibrosis and metabolic decline. Cell Metab. 36,
Med. 25, 1822–1832. https://doi.org/10.1038/s41591-019-0675-0. 793–807.e5. https://doi.org/10.1016/j.cmet.2024.01.015.
108. Li, Y., Wu, A., Jin, X., Shen, H., Zhao, C., Yi, X., Nie, H., Wang, M., Yin, S., 125. Wang, S., Zheng, Y., Li, J., Yu, Y., Zhang, W., Song, M., Liu, Z., Min, Z.,
Zuo, H., et al. (2024). DDO1002, an NRF2-KEAP1 inhibitor, improves he- Hu, H., Jing, Y., et al. (2020). Single-Cell Transcriptomic Atlas of Primate
matopoietic stem cell aging and stress response. Life Med. 3, lnae043. Ovarian Aging. Cell 180, 585–600.e19. https://doi.org/10.1016/j.cell.
https://doi.org/10.1093/lifemedi/lnae043. 2020.01.009.
109. Sonninen, T.M., Goldsteins, G., Laham-Karam, N., Koistinaho, J., and 126. Du Clos, T.W., and Mold, C. (2011). Pentraxins (CRP, SAP) in the process
� (2020). Proteostasis Disturbances and Inflammation in
Lehtonen, S. of complement activation and clearance of apoptotic bodies through Fcγ
Neurodegenerative Diseases. Cells 9, 2183. https://doi.org/10.3390/ receptors. Curr. Opin. Organ Transplant. 16, 15–20. https://doi.org/10.
cells9102183. 1097/MOT.0b013e32834253c7.
110. Korovila, I., Hugo, M., Castro, J.P., Weber, D., Höhn, A., Grune, T., and 127. Geng, L., Ping, J., Wu, R., Yan, H., Zhang, H., Zhuang, Y., Ning, T., Wang,
Jung, T. (2017). Proteostasis, oxidative stress and aging. Redox Biol. J., Liang, C., Zhang, J., et al. (2025). Systematic profiling reveals betaine
13, 550–567. https://doi.org/10.1016/j.redox.2017.07.008. as an exercise mimetic for geroprotection. Cell. https://doi.org/10.1016/j.
cell.2025.06.001.
111. Jiménez-Loygorri, J.I., and Boya, P. (2024). Aging STINGs: mitophagy at
128. Sun, G., Li, K., Ping, J., Zhao, L., Cui, C., Wu, J., Xie, L., Yao, X., Xu, G.,
the crossroads of neuroinflammation. Autophagy 20, 1684–1686. https://
Ma, S., et al. (2025). A single-cell transcriptomic atlas reveals senescence
doi.org/10.1080/15548627.2024.2322421.
and inflammation in the post-tuberculosis human lung. Nat. Microbiol.
112. Franceschi, C., Garagnani, P., Parini, P., Giuliani, C., and Santoro, A. https://doi.org/10.1038/s41564-025-02050-3.
(2018). Inflammaging: a new immune-metabolic viewpoint for age-
129. Wu, R., Sun, F., Zhang, W., Ren, J., and Liu, G.H. (2024). Targeting aging
related diseases. Nat. Rev. Endocrinol. 14, 576–590. https://doi.org/10.
and age-related diseases with vaccines. Nat Aging 4, 464–482. https://
1038/s41574-018-0059-4.
doi.org/10.1038/s43587-024-00597-0.
113. Yan, H., Jiang, M., Qu, J., and Liu, G.-H. (2025). IgG: An Emerging Hall-
130. Schneider, C.A., Rasband, W.S., and Eliceiri, K.W. (2012). NIH Image to
mark of Aging. Oral Sci. Homeost. Med. 1. https://doi.org/10.26599/
ImageJ: 25 years of image analysis. Nat. Methods 9, 671–675. https://
OSHM.2025.9610001.
doi.org/10.1038/nmeth.2089.
114. Wechalekar, A.D., Gillmore, J.D., and Hawkins, P.N. (2016). Systemic 131. Demichev, V., Messner, C.B., Vernardis, S.I., Lilley, K.S., and Ralser, M.
amyloidosis. Lancet 387, 2641–2654. https://doi.org/10.1016/S0140- (2020). DIA-NN: neural networks and interference correction enable deep
6736(15)01274-X. proteome coverage in high throughput. Nat. Methods 17, 41–44. https://
115. Westermark, G.T., Fändrich, M., and Westermark, P. (2015). AA amyloid- doi.org/10.1038/s41592-019-0638-x.
osis: pathogenesis and targeted therapy. Annu. Rev. Pathol. 10, 132. Feng, Z., Fang, P., Zheng, H., and Zhang, X. (2023). DEP2: an upgraded
321–344. https://doi.org/10.1146/annurev-pathol-020712-163913. comprehensive analysis toolkit for quantitative proteomics data. Bioin-
116. Gertz, M.A., Dispenzieri, A., and Sher, T. (2015). Pathophysiology and formatics 39, btad526. https://doi.org/10.1093/bioinformatics/btad526.
treatment of cardiac amyloidosis. Nat. Rev. Cardiol. 12, 91–102. 133. Hao, Z., Lv, D., Ge, Y., Shi, J., Weijers, D., Yu, G., and Chen, J. (2020).
https://doi.org/10.1038/nrcardio.2014.165. RIdeogram: drawing SVG graphics to visualize and map genome-wide
ll
Article
data on the idiograms. PeerJ Comput. Sci. 6, e251. https://doi.org/10. human synovium through single-nucleus transcriptomic profiling. Protein
7717/peerj-cs.251. Cell 15, 441–459. https://doi.org/10.1093/procel/pwad060.
134. Ritchie, M.E., Phipson, B., Wu, D., Hu, Y., Law, C.W., Shi, W., and Smyth, 152. Yang, S., Liu, C., Jiang, M., Liu, X., Geng, L., Zhang, Y., Sun, S., Wang, K.,
G.K. (2015). limma powers differential expression analyses for RNA- Yin, J., Ma, S., et al. (2024). A single-nucleus transcriptomic atlas of pri-
sequencing and microarray studies. Nucleic Acids Res. 43, e47. mate liver aging uncovers the pro-senescence role of SREBP2 in hepa-
https://doi.org/10.1093/nar/gkv007. tocytes. Protein Cell 15, 98–120. https://doi.org/10.1093/procel/
135. Wickham, H. (2016). ggplot2: Elegant Graphics for Data Analysis (New pwad039.
York: Springer-Verlag). https://doi.org/10.1007/978-3-319-24277-4. 153. Hu, Q., Zhang, B., Jing, Y., Ma, S., Hu, L., Li, J., Zheng, Y., Xin, Z., Peng,
136. Gu, Z. (2022). Complex heatmap visualization. Imeta 1, e43. https://doi. J., Wang, S., et al. (2024). Single-nucleus transcriptomics uncovers a ger-
org/10.1002/imt2.43. oprotective role of YAP in primate gingival aging. Protein Cell 15,
137. Shannon, P., Markiel, A., Ozier, O., Baliga, N.S., Wang, J.T., Ramage, D., 612–632. https://doi.org/10.1093/procel/pwae017.
Amin, N., Schwikowski, B., and Ideker, T. (2003). Cytoscape: a software 154. Huang, D., Zuo, Y., Zhang, C., Sun, G., Jing, Y., Lei, J., Ma, S., Sun, S.,
environment for integrated models of biomolecular interaction networks. Lu, H., Cai, Y., et al. (2023). A single-nucleus transcriptomic atlas of pri-
Genome Res. 13, 2498–2504. https://doi.org/10.1101/gr.1239303. mate testicular aging reveals exhaustion of the spermatogonial stem cell
138. Lehallier, B., Gate, D., Schaum, N., Nanasi, T., Lee, S.E., Yousef, H., reservoir and loss of Sertoli cell homeostasis. Protein Cell 14, 888–907.
Moran Losada, P., Berdnik, D., Keller, A., Verghese, J., et al. (2019). Un- https://doi.org/10.1093/procel/pwac057.
dulating changes in human plasma proteome profiles across the lifespan. 155. Lei, J., Jiang, X., Huang, D., Jing, Y., Yang, S., Geng, L., Yan, Y., Zheng,
Nat. Med. 25, 1843–1850. https://doi.org/10.1038/s41591-019-0673-2. F., Cheng, F., Zhang, W., et al. (2024). Human ESC-derived vascular cells
139. Dobin, A., Davis, C.A., Schlesinger, F., Drenkow, J., Zaleski, C., Jha, S., promote vascular regeneration in a HIF-1α dependent manner. Protein
Batut, P., Chaisson, M., and Gingeras, T.R. (2013). STAR: ultrafast uni- Cell 15, 36–51. https://doi.org/10.1093/procel/pwad027.
versal RNA-seq aligner. Bioinformatics 29, 15–21. https://doi.org/10. 156. Uras, F., Küçük, B., Bingöl Özakpınar, Ö., and Demir, A.M. (2015). Growth
1093/bioinformatics/bts635. Arrest-Specific 6 (Gas6) and TAM Receptors in Mouse Platelets. Turk. J.
140. Liao, Y., Smyth, G.K., and Shi, W. (2014). featureCounts: an efficient gen- Haematol. 32, 58–63. https://doi.org/10.4274/tjh.2013.0097.
eral purpose program for assigning sequence reads to genomic features. 157. Rejas-González, R., Montero-Calle, A., Valverde, A., Salvador, N.P., Car-
Bioinformatics 30, 923–930. https://doi.org/10.1093/bioinformatics/ ballés, M.J.C., Ausı́n-González, E., Sánchez-Naves, J., Campuzano, S.,
btt656. Barderas, R., and Guzman-Aranguez, A. (2024). Proteomics Analyses
141. Love, M.I., Huber, W., and Anders, S. (2014). Moderated estimation of of Small Extracellular Vesicles of Aqueous Humor: Identification and Vali-
fold change and dispersion for RNA-seq data with DESeq2. Genome dation of GAS6 and SPP1 as Glaucoma Markers. Int. J. Mol. Sci. 25,
Biol. 15, 550. https://doi.org/10.1186/s13059-014-0550-8. 6995. https://doi.org/10.3390/ijms25136995.
142. Wu, T., Hu, E., Xu, S., Chen, M., Guo, P., Dai, Z., Feng, T., Zhou, L., Tang, 158. Belgiovine, C., Digifico, E., Erreni, M., Putignano, A.R., Mannarino, L.,
W., Zhan, L., et al. (2021). clusterProfiler 4.0: A universal enrichment tool Valentino, S., Grizzi, F., Pasqualini, F., Recordati, C., Bertola, L., et al.
for interpreting omics data. Innovation (Camb.) 2, 100141. https://doi. (2025). Malignant mesothelioma-associated inflammatory microenviron-
org/10.1016/j.xinn.2021.100141. ment promotes tumor progression via GPNMB. J. Transl. Med. 23, 454.
143. Hänzelmann, S., Castelo, R., and Guinney, J. (2013). GSVA: gene set https://doi.org/10.1186/s12967-025-06407-4.
variation analysis for microarray and RNA-seq data. BMC Bioinformatics 159. Fukaura, M., Ishitsuka, Y., Shirakawa, S., Ushihama, N., Yamada, Y.,
14, 7. https://doi.org/10.1186/1471-2105-14-7. Kondo, Y., Takeo, T., Nakagata, N., Motoyama, K., Higashi, T., et al.
144. Pedregosa, F., Varoquaux, G., Gramfort, A., Michel, V., Thirion, B., Grisel, (2021). Intracerebroventricular Treatment with 2-Hydroxypropyl-β--
O., Blondel, M., Prettenhofer, P., Weiss, R., Dubourg, V., et al. Scikit- Cyclodextrin Decreased Cerebellar and Hepatic Glycoprotein Nonmeta-
learn: Machine Learning in Python. J. Mach. Learn. Res. 12, 2825–2830. static Melanoma Protein B (GPNMB) Expression in Niemann-Pick Dis-
145. Peng, Y., Ding, L., Xiao, Z., Song, M., Lv, J., and Liu, G.H. (2024). Ethical ease Type C Model Mice. Int. J. Mol. Sci. 22, 452. https://doi.org/10.
concerns in aging research: perspectives of global frontline researchers. 3390/ijms22010452.
Sci. China Life Sci. 67, 2149–2156. https://doi.org/10.1007/s11427-024- 160. Eskes, E.C.B., van der Lienden, M.J.C., Sjouke, B., van Vliet, L., Brands,
2650-y. M., Hollak, C.E.M., and Aerts, J. (2023). Glycoprotein non-metastatic
146. Peng, Y., Ding, L., Song, M., Xiao, Z., Lv, J., and Liu, G.H. (2023). Acting protein B (GPNMB) plasma values in patients with chronic visceral acid
on ethics and governance of aging research. Trends Mol. Med. 29, sphingomyelinase deficiency. Mol. Genet. Metab. 139, 107631. https://
419–421. https://doi.org/10.1016/j.molmed.2023.03.004. doi.org/10.1016/j.ymgme.2023.107631.
147. Zhang, W., Zhang, S., Yan, P., Ren, J., Song, M., Li, J., Lei, J., Pan, H., 161. Sainaghi, P.P., Alciato, F., Carnieletto, S., Castello, L., Bergamasco, L.,
Wang, S., Ma, X., et al. (2020). A single-cell transcriptomic landscape Sola, D., Bongo, A.S., Inglese, E., Polosa, R., and Avanzi, G.C. (2009).
of primate arterial aging. Nat. Commun. 11, 2202. https://doi.org/10. Gas6 evaluation in patients with acute dyspnea due to suspected pulmo-
1038/s41467-020-15997-0. nary embolism. Respir. Med. 103, 589–594. https://doi.org/10.1016/j.
148. Patel, J.J., Srivastava, S., and Siow, R.C.M. (2016). Isolation, Culture, rmed.2008.10.018.
and Characterization of Vascular Smooth Muscle Cells. Methods Mol. 162. Sun, S., Ma, S., Cai, Y., Wang, S., Ren, J., Yang, Y., Ping, J., Wang, X.,
Biol. 1430, 91–105. https://doi.org/10.1007/978-1-4939-3628-1_6. Zhang, Y., Yan, H., et al. (2023). A single-cell transcriptomic atlas of ex-
149. Niu, L., Stinson, S.E., Holm, L.A., Lund, M.A.V., Fonvig, C.E., Cobuccio, ercise-induced anti-inflammatory and geroprotective effects across the
L., Meisner, J., Juel, H.B., Fadista, J., Thiele, M., et al. (2025). Plasma pro- body. Innovation (Camb.) 4, 100380. https://doi.org/10.1016/j.xinn.
teome variation and its genetic determinants in children and adolescents. 2023.100380.
Nat. Genet. 57, 635–646. https://doi.org/10.1038/s41588-025-02089-2. 163. Diamanti, K., Cavalli, M., Pereira, M.J., Pan, G., Castillejo-López, C., Ku-
150. Koh, B., Liu, M., Almonte, R., Ariad, D., Bundalian, G., Chan, J., Choi, J., mar, C., Mundt, F., Komorowski, J., Deshmukh, A.S., Mann, M., et al.
Chou, W.-F., Cuaresma, R., et al. (2024). Multi-omics profiling with untar- (2022). Organ-specific metabolic pathways distinguish prediabetes,
geted proteomics for blood-based early detection of lung cancer. Pre- type 2 diabetes, and normal tissues. Cell Rep. Med. 3, 100763. https://
print at medRxiv. https://doi.org/10.1101/2024.01.03.24300798. doi.org/10.1016/j.xcrm.2022.100763.
151. Liu, F., Lu, Y., Wang, X., Sun, S., Pan, H., Wang, M., Wang, Z., Zhang, W., 164. Wei, R., Wang, J., Su, M., Jia, E., Chen, S., Chen, T., and Ni, Y. (2018).
Ma, S., Sun, G., et al. (2024). Identification of FOXO1 as a geroprotector in Missing Value Imputation Approach for Mass Spectrometry-based
ll
Article
Metabolomics Data. Sci. Rep. 8, 663. https://doi.org/10.1038/s41598- 167. Love, M.I., Huber, W., and Anders, S. (2014). Moderated estimation of
017-19120-0. fold change and dispersion for RNA-seq data with DESeq2. Genome
Biol. 15, 550. https://doi.org/10.1186/s13059-014-0550-8.
165. Frankish, A., Diekhans, M., Jungreis, I., Lagarde, J., Loveland, J.E., 168. Marakulina, D., Vorontsov, I.E., Kulakovskiy, I.V., Lennartsson, A.,
Mudge, J.M., Sisu, C., Wright, J.C., Armstrong, J., Barnes, I., et al. Drabløs, F., and Medvedeva, Y.A. (2023). EpiFactors 2022: expansion
(2021). GENCODE 2021. Nucleic Acids Res. 49, D916–D923. https:// and enhancement of a curated database of human epigenetic factors
doi.org/10.1093/nar/gkaa1087. and complexes. Nucleic Acids Res. 51, D564–D570. https://doi.org/10.
1093/nar/gkac989.
166. Uhlén, M., Fagerberg, L., Hallström, B.M., Lindskog, C., Oksvold, P., 169. Steinkamp, R., Tsitsiridis, G., Brauner, B., Montrone, C., Fobo, G., Frish-
Mardinoglu, A., Sivertsson, Å., Kampf, C., Sjöstedt, E., Asplund, A., man, G., Avram, S., Oprea, T.I., and Ruepp, A. (2025). CORUM in 2024:
et al. (2015). Proteomics. Tissue-based map of the human proteome. Sci- protein complexes as drug targets. Nucleic Acids Res. 53, D651–D657.
ence 347, 1260419. https://doi.org/10.1126/science.1260419. https://doi.org/10.1093/nar/gkae1033.
ll
Article
STAR★METHODS
ll
Article
Continued
REAGENT or RESOURCE SOURCE IDENTIFIER
Alexa Fluor 647 Donkey Anti-Rabbit IgG (H+L) Invitrogen Cat# A-31573; RRID: AB_2536183
Alexa Fluor 488 Donkey Anti-Mouse IgG (H+L) Invitrogen Cat# A-21202; RRID: AB_141607
Alexa Fluor 568 Donkey Anti-Mouse IgG (H+L) Invitrogen Cat# A-10037; RRID: AB_2534013
Alexa Fluor(R) 488 anti-mouse MAdCAM-1 BioLegend Cat# A-120708; RRID: AB_493398
Alexa Fluor 488 Donkey Anti-Rat IgG (H+L) Invitrogen Cat# A-21208; RRID: AB_2535794
Biological samples
Human heart, aorta, lung, muscle, spleen, West China Hospital of N/A
lymph, adrenal, skin, liver, intestine, Sichuan University
pancreas, adipose and peripheral
blood for proteomics analysis
Human peripheral blood for ELISA analysis Quzhou People’s Hospital, N/A
Quzhou, China
Experimental models: Organisms/strains
C57BL/6J male mice Specific Pathogen Free (SPF) N/A
Biotechnology Co., Ltd.,
Beijing, China
Experimental models: Cell lines
Human aortic endothelial cells Lonza Cat# CC-2535
Human vascular smooth muscle cells This study, see STAR Methods N/A
Human acute monocytic leukemia cells (THP-1) Pricella Cat# CL-0233
Chemicals, peptides, and recombinant proteins
Hoechst 33342 Thermo Fisher Scientific Cat# H3570; RRID: N/A
VECTASHIELD Antifade Mounting Medium Neobioscience Cat# H-1000
Calcein AM Invitrogen Cat# C3099
EBM®-2 Basal Medium Lonza Cat# CC-3156
EGM®-2 Endothelial SingleQuots® Kit Lonza Cat# CC-4176
Fetal bovine serum GEMINI Cat# 900-108
DMEM/High Glucose HyClone Cat# SH30243.01
Accumax Millipore Cat# SCR006
Accutase Sigma Cat# A6964
Penicillin/streptomycin (P/S) GIBCO Cat# 15140-163
TrypLE Express Enzyme (1×), no phenol red GIBCO Cat# 12563029
HiScript III RT SuperMix for qPCR (+gDNA wiper) Vazyme Cat# R323-01
NovoStart®SYBR qPCR SuperMix Plus Novoprotein Cat# E096-01B
4% paraformaldehyde, PFA Dingguo, China Cat# AR-0211
Triton X-100 Sigma-Aldrich Cat# T9284
Trypsin Promega Cat# V5113
Protease Inhibitor Cocktail Merck Millipore Cat# P8340
Tetraethylammonium bromide Sigma Cat# TX0255
Trichloroacetic acid Sigma Cat# 822342
DL-Dithiothreitol Sigma Cat# LEYH9ACF09F3
Iodoacetamide Sigma Cat# I1149
Protease inhibitor cocktail Roche Cat# 4693159001
TRIzol™ Reagent Thermo Fisher Scientific Cat# 15596018
NP-40 Dingguo, China Cat# DH218
pH 6.0 sodium citrate buffer ZSGB-BIO Cat# ZLI-9064
Normal Donkey Serum Jackson ImmunoResearch Cat# 017-000-121; RRID: AB_2337258
X-Gal Amresco Cat# 0428-25G
(Continued on next page)
ll
Article
Continued
REAGENT or RESOURCE SOURCE IDENTIFIER
PVDF membranes Millipore Cat# IPVH00010
GAS6 recombinant protein MCE Cat# HY-P70780
SLPI recombinant protein MCE Cat# HY-P73414
HTRA1 recombinant protein MCE Cat# HY-P701005
COMP recombinant protein MCE Cat# HY-P72945
IGFBP7 recombinant protein MCE Cat# HY-P70726
NOTCH3 recombinant protein MCE Cat# HY-P78009
GPNMB recombinant protein MCE Cat# HY-P75951
NEGR1 recombinant protein MCE Cat# HY-P76508
SAP recombinant protein MCE Cat# HY-P71067
Recombinant mouse GPNMB protein MCE Cat# HY-P77116
Recombinant mouse GAS6 protein MCE Cat# HY-P75174A
Critical commercial assays
Top 14 Abundant Protein Thermo Fisher Scientific Cat# A36371
Depletion Spin Columns Kit
BCA Protein Assay Kit Dingguo, China Cat# BCA02
DAB Staining Kit ZSGB-BIO Cat# ZLI-9018; PV-9001; PV-9002
Human HTRA1 ELISA Kit Elabscience Cat# E-EL-H6300
Human CHI3L1 ELISA Kit CUSABIO Cat# CSB-E13608h
Human NOTCH3 ELISA Kit CUSABIO Cat# CSB-EL015952HU
Human EFEMP1 ELISA Kit CUSABIO Cat# CSB-EL007450HU
Human LTBP2 ELISA Kit CUSABIO Cat# CSB-E11268h
Human ITLN1 ELISA Kit Elabscience Cat# E-EL-H2028
Human COMP ELISA Kit Elabscience Cat# E-EL-H0654
Human IL-6 ELISA Kit BioLegend Cat# 430515
Human GAS6 ELISA Kit Elabscience Cat# E-EL-H6014
Human HARS ELISA Kit CUSABIO Cat# CSB-EL010137HU
Deposited data
The raw proteomic data This study IPX0010296000
The raw bulk RNA-seq data This study GSA: HRA009355, HRA011245
Oligonucleotides
Primers for qRT-PCR, see Table S5 This paper N/A
Software and algorithms
GraphPad Prism (version 8.0.2) GraphPad Software Inc. https://www.graphpad.com/
Image J Schneider et al.130 https://imagej.nih.gov/ij/
DIA-NN (version 1.8) Demichev et al.131 https://github.com/vdemichev/DiaNN
DEP2 (version 0.5.4.02) Feng et al.132 https://github.com/mildpiggy/DEP2
RIdeogram (version 0.2.2) Hao et al.133 https://cran.r-project.org/web/
packages/RIdeogram/vignettes/
RIdeogram.html
limma (version 3.58.1) Ritchie et al.134 https://bioconductor.org/packages/
release/bioc/html/limma.html
ggplot2 (version 3.4.2) Wickham et al.135 https://ggplot2.tidyverse.org/
ComplexHeatmap (version 2.18.0) Gu136 https://github.com/jokergoo/ComplexHeatmap
Cytoscape (version 3.10.2) Shannon et al.137 https://cytoscape.org/
DEswan Benoit et al.138 https://github.com/lehallib/DEswan
Trim Galore (version 0.6.10) N/A https://github.com/FelixKrueger/TrimGalore
STAR (version 2.7.11a) Dobin et al.139 https://github.com/alexdobin/STAR
(Continued on next page)
ll
Article
Continued
REAGENT or RESOURCE SOURCE IDENTIFIER
140
featureCounts (version 2.0.6) Liao et al. https://subread.sourceforge.net/
featureCounts.html
DESeq2 (version 1.44.0) Love et al.141 https://github.com/mikelove/DESeq2
pheatmap (version 1.0.12) N/A https://cran.r-project.org/web/packages/
pheatmap/index.html
clusterProfiler (version 4.10.0) Wu et al.142 https://bioconductor.org/packages/
release/bioc/html/clusterProfiler.html
GSVA (version 1.50.0) Hänzelmann et al.143 https://bioconductor.org/packages/
devel/bioc/vignettes/GSVA/inst/
doc/GSVA.html
glmnet (version 4.1_8) N/A https://doi.org/10.18637/jss.v033.i01
mlr3verse (version 0.2.8) N/A https://github.com/mlr-org/mlr3verse
iml (version 0.11.1) N/A https://github.com/giuseppec/iml
CellPhoneDB (version 5.0.1) Garcia-Alonso et al.95 https://github.com/ventolab/CellphoneDB
Tidygraph (version 1.2.3) N/A https://github.com/thomasp85/tidygraph
Scikit-learn (version 1.3.0) Pedregosa et al.144 https://github.com/scikit-learn/scikit-learn
Ethical statements
This study adheres to the guiding principles of the Declaration of Helsinki and is conducted in accordance with ethical princi-
ples.145,146 Prior approval was obtained for the use of human genetic resources from the Ethics Committee of West China Hospital,
Sichuan University (NO. 2023-801), Quzhou People’s Hospital (2020-12-001), Beijing Institute of Genomics (China National Center for
Bioinformation) of the Chinese Academy of Sciences (NO. 2023H045) and Beijing Anzhen hospital (NO. 2021-28). All mouse exper-
imental procedures in this study were conducted in accordance with the guidelines set by the Animal Care and Use Institutional Com-
mittee of the Institute of Zoology, Chinese Academy of Sciences (IOZ-IACUC-2025-215).
Human samples
A total of 76 organ transplant donors (aged 14-68 years, 57 males and 19 females) who suffered accidental traumatic brain injury were
included. All samples in this study were obtained from individuals of Chinese ethnicity. Human tissue donors were not subjected to
any experimental procedures prior to donation. All samples were collected postmortem following the standard organ procurement
procedures. Prior to sample collection, detailed explanations regarding the purpose and processing procedures of the samples were
provided to the donors’ families, and written informed consent was obtained. The plasma samples for the independent validation of
ELISA experiments were sourced from a separate cohort in Quzhou.24
Sample size was determined based on data availability and constraints of human tissue acquisition. Given the exploratory nature of
multi-organ human proteomic profiling, formal statistical power calculations were not feasible. Nonetheless, we included biological
replicates for each tissue and age group to ensure representation across decades for trajectory analyses. All human tissue and
plasma samples were grouped based on chronological age according to donor metadata. Detailed sample information was summa-
rized in Table S1.
Cell culture
Human aortic endothelial cells (hAECs) were purchased from Lonza (Lonza, CC-2535), and cultured in accordance with recommen-
ded standard endothelial medium, containing EBM®-2 Basal Medium (Lonza, CC-3156) and EGM®-2 MV Microvascular Endothelial
Cell Growth Medium SingleQuots® supplements (FBS, Hydrocortisone, hFGF-B, VEGF, R3-IGF-1, Ascorbic Acid, hEGF, GA-1000
and Heparin) (Lonza, CC-4176). Endothelial cells are persistently cultured in Advanced BioMatrix-precoated collagen dishes, with the
fresh culture medium replaced every 48 hours to maintain an optimal environment for cell growth. Digestion of cells is performed us-
ing Accumax (Millipore), and passaging is conducted when the cell confluence reaches 80%.147
ll
Article
Human vascular smooth muscle cells (hVSMCs) were isolated as follows.148 Tunica intima was scratched from the aorta, and
tunica media was reserved for hVSMCs isolation. The tunica media was diced into 0.5 mm-sized fragments, supplemented with
0.25% trypsin in a 37◦ C water-bath for 3.5 hours. This enzymatic digestion process was ceased by adding an equal volume of
DMEM medium. The mixture was passed through a 120 μm sterilized nylon mesh filter (Millipore) to remove undigested debris.
The strained solution was centrifuged at 500 rpm for five minutes, after which the pellet was reconstituted and cultured in recommen-
ded culturing medium DMEM medium (DMEM, HyClone) supplemented with 10% fetal bovine serum (FBS, GEMINI), 2 mM
GlutaMAXTM (Gibco), 0.1 mM MEM non-essential amino acids (NEAA, Gibco) and 1% penicillin/streptomycin (Gibco). Fresh culture
medium was replaced every 48 hours to maintain an optimal environment for cell growth. Cell passaging was conducted when the
cell confluence reached 80%.
The THP-1 cells were purchased from Pricella (Pricella) and cultured in DMEM/High Glucose (HyClone) medium supplemented
with 10% FBS (GEMINI), 1% penicillin/streptomycin (Gibco), 0.1 mM NEAA (Gibco) and 2 mM GlutMaxTM (Gibco). The cells were
centrifuged every 48 hours to remove the spent medium and replace it with fresh medium to maintain the optimal growth
environment.
METHOD DETAILS
ll
Article
reduction. Subsequently, iodoacetamide (IAA) was added to 11 mM final concentration and incubated at room temperature in the
dark for 15 minutes for alkylation.
LC-MS/MS Analysis
Peptides were dissolved in mobile phase A and separated using the Vanquish Neo ultra-high performance liquid chromatography
(UHPLC) system. Mobile phase A consisted of an aqueous solution containing 0.1% formic acid, while mobile phase B was an
aqueous solution containing 0.1% formic acid and 80% acetonitrile. The liquid chromatography gradient for plasma was set as fol-
lows: 0-1.6 minutes, 4%-22.5% B; 1.6-2.0 minutes, 22.5%-35% B; 2.0-2.6 minutes, 35%-55% B; 2.6-2.7 minutes, 55%-99% B;
2.7-6.8 minutes, 99% B; 6.8-7.6 minutes, 99% B, with a flow rate maintained at 300 nL/minutes. For tissues other than plasma,
the gradient was as follows: 0-0.5 minutes, 4% B; 0.5-0.6 minutes, 4%-8% B; 0.6-13.6 minutes, 8%-22.5% B; 13.6-20.5 minutes,
22.5%-35% B; 20.5-20.9 minutes, 35%-55% B; 20.9-21.4 minutes, 55%-99% B; 21.4-22.6 minutes, 99% B, with a flow rate main-
tained at 400 nL/minutes. After separation by the UHPLC system, the peptides were introduced into the NSI ion source for ionization,
followed by analysis using the Orbitrap Astral mass spectrometer. The ion source voltage was set at 1,900 V, with parent peptide ions
detected and analyzed by the Orbitrap detector, and the secondary fragment ions detected by the Astral detector. The primary mass
spectrometry scan range was set from 380 to 980 m/z, with a scanning resolution of 240,000; the secondary mass spectrometry scan
range was fixed at a starting point of 150 m/z, with a scanning resolution of 80,000. Data acquisition was conducted in data-inde-
pendent acquisition (DIA) mode, where multiple continuous m/z windows followed the primary scan, and peptide ions were intro-
duced into the higher-energy collisional dissociation (HCD) collision pool with 25% fragmentation energy for collision-induced disso-
ciation, followed by secondary mass spectrometry analysis. To improve the mass spectrometry efficiency, the automatic gain control
(AGC) was set to 500%, and the maximum injection time was set to 3 milliseconds.
Database search
The raw data from the mass spectrometer were imported into the search software, and the DIA-NN (version 1.8) search engine was
used with default parameters. The database used was Homo_sapiens_9606_SP_20230103.fasta (20,389 sequences), with ‘‘trypsin/
P’’ as the cleavage enzyme and a maximum missed cleavage set to 1. Fixed modifications were set to: ‘‘N-term M excision’’ and ‘‘C
carbamidomethylation’’. A theoretical spectral library was constructed using deep learning algorithms, and a decoy database was
included to calculate the false discovery rate (FDR) caused by random matches. The FDR for precursor identification was set at
1%. To obtain high-quality results, the search results were further filtered with the criteria set as follows: precursor and protein
FDR were both set to 1%; proteins must be identified with at least one peptide.
ELISA assay
The ELISA experiment was performed following the standard protocol of the reference kit. To put it succinctly, plasma samples with
EDTA as an anticoagulant were gently thawed on ice and centrifuged. The resulting supernatant was then diluted according to the
recommended ratio and, along with the standards, was added to an ELISA plate that has been pre-coated with antibodies or antigens
for incubation. A sequence of reagents including biotinylated antibodies, horseradish peroxidase-conjugated streptavidin, TMB sub-
strate solution, and stopping solution was added, with the reaction proceeding for an appropriate time. Absorbance was measured at
450 nm, and the standard curve was constructed using the recommended professional software Curve Expert to determine the sam-
ple concentration. The statistical analysis was conducted with GraphPad (version 8.0.2) software. The plasma samples used for
ELISA are detailed in Table S1.
Western blotting
Western blotting assay was performed with reference to published procedures.78,153 For the processing of tissue samples, retrieved
the tissue samples from liquid nitrogen, wrapped them in aluminum foil to prevent splashing, and ground them into a fine powder
using a mortar and pestle. Added RIPA lysis buffer and lysed at 4◦ C for 1 hour. After centrifugation, the supernatant was collected.
ll
Article
For cell samples, washed the adherent cultured cells with PBS, then added 1×SDS protein lysis buffer and lysed on ice for 30 minutes,
collecting the lysate. Boiled the aforementioned lysate at 95◦ C for 5 minutes to denature the proteins. Quantified the proteins using a
BCA assay kit, and separated proteins of different molecular weights by SDS-PAGE gel electrophoresis. Subsequently, the samples
were incubated with specific primary antibodies and horseradish peroxidase (HRP)-labeled secondary antibodies, and collected the
protein luminescent signals using a Bio-Rad system. The protein grayscale was analyzed by using Image J software, and statistical
analysis and presentation were carried out with GraphPad (version 8.0.2) software.
SA-β-Gal staining
SA-β-Gal staining assay was performed with reference to published procedures.77,154 Briefly, after rinsing with PBS, adherent cells
were treated with fixative solution (2% formaldehyde and 0.2% glutaraldehyde solution) and left to stand for 5 minutes at room tem-
perature to fix the cells. Subsequently, X-Gal staining solution was added under a pH 6.0 buffer system and incubated in the dark at
37◦ C for an appropriate time. Representative image capture was performed using the microscope system (Nikon, Y-TV55), and the
proportion of SA-β-Gal positive cells was calculated using Image J software, with statistical analysis conducted using GraphPad
(version 8.0.2) software.
ll
Article
BODIPY staining
Frozen tissue sections (10 μm thick) were air-dried for 15 minutes, washed three times with PBS, fixed with 4% PFA for 10 minutes,
permeabilized with 0.4% Triton X-100 for 30 minutes, and incubated with primary antibodies overnight. During secondary antibody
incubation, 10 μg/mL BODIPY was added and incubated at room temperature for 1 hour. The tissues were stained with Hoechst
33342 and mounted using anti-fade mounting medium. Images were acquired using the Zeiss LSM 980 confocal microscope system.
ll
Article
Classification of proteins based on subcellular location, cellular component, and molecular function
The subcellular location information of each protein was derived from the predicted protein class obtained from the Human Protein
Atlas (https://www.proteinatlas.org/).166 Gene sets of different cellular components and molecular functions were obtained from the
Molecular Signatures Database (MSigDB) (https://www.gsea-msigdb.org/gsea/msigdb). The number and proportion of proteins
belonging to different categories across tissues were calculated and visualized using the ggplot2 package (version 3.4.2).
ll
Article
ll
Article
age-group-specific DEPs and lifespan-wide DEPs. This analysis utilized the clusterProfiler R package (version 4.10.0) within R soft-
ware (version 4.3.3).142
ll
Article
All statistical analyses and data handling were performed using GraphPad Prism software (version 8.0.2). Data are presented
as mean ± standard error of the mean (SEM). Statistical differences between two groups were assessed using two-tailed t-tests
(passed normality test) or Mann-Whitney test (not passed normality test), whereas those among more than two groups were as-
sessed using one-way analysis of variance (ANOVA). In histological staining analysis, fold-change data were calculated relative to
the mean of all samples, and Spearman’s rank correlation test was used to calculate the correlation coefficient and p values of
age-related data. In plasma ELISA experiments, Pearson correlation analysis was used to calculate the correlation coefficient and
p values of age-related data. Significant differences between groups are indicated either by p values directly in the figure or by as-
terisks: * p value < 0.05, ** p value < 0.01, *** p value < 0.001.
Supplemental figures
Figure S1. Gender, tissue bias, and functional attributes of identified proteins, related to Figure 1
(A) Histograms of protein abundance distribution in each tissue. Red: before quantile regression imputation of left-censored data (QRILC) imputation. Blue: after
QRILC imputation.
(B) UMAP plot showing the distribution of samples across different tissues.
(C) Heatmap of Spearman’s correlation coefficients between protein expression levels in each tissue. Color key: white to red (low to high correlation).
(D) Left: pie charts of protein proportions in different categories across tissues. Right: bar plot of ‘‘tissue-enriched’’ proteins.
(E) Left: heatmap of tissue-enriched protein expression across tissues. Right: representative GO terms enriched in tissue-enriched proteins. Color key: white to
red (− log10(p value) low to high).
(F) Western blot of tissue-specific proteins, with Ponceau S as a loading control. Left: protein structures predicted using AlphaFold.
(G) Left: heatmap of tissue-enhanced protein expression across tissues, categorized into modules. Right: representative GO terms enriched in the top 10
modules. Color key: blue to red (scaled expression level low to high).
(H) Left: heatmap of ‘‘housekeeping’’ and ‘‘others’’ protein expression across tissues. Right: representative GO terms enriched in these categories. Color key:
blue to red (protein abundance low to high).
(I) Radar chart of sex-biased protein counts across tissues.
(J) Top: pie charts of sex-biased proteins (p < 0.05, |log2FC| > 2) across tissues. Bottom: scatterplot of sex-biased protein differences between females and males,
with top 2 labeled.
(K) Western blot of proteins with sex biases: SGF29 (female biases in heart) and MMP3 (male biases in adipose). Ponceau S as loading control.
ll
Article
Figure S2. Aging-related mRNA-protein decoupling and proteostasis loss, related to Figure 1
(A) Schematic of protein homeostasis maintenance. Protein quality control occurs at transcription, translation, folding, modification, transport, localization, and
function as complexes. Proteostasis ensures normal tissue function and specificity and contributes to sex-based differences.
(B) Density plot of Spearman’s correlation loss between protein abundance and mRNA expression during aging. Blue: young group; red: old group.
(C) Sankey plot of decoupling proteins showing loss of Spearman’s correlation between protein abundance and mRNA expression in the aged group. Red:
positive correlation; blue: negative correlation; light gray: other proteins.
(D) Bar plot of GO terms for decoupling genes in each tissue. Color: white to red (− log10(p value) low to high).
(E) x axis: age-related correlation; y axis: decoupling difference. Red: age-positively correlated decoupling proteins. Blue: age-negatively correlated decoupling
proteins. Gray: other decoupling proteins.
(F) Dot plot of GSEA for protein homeostasis pathways for DEPs. Dot size: − log10(p value); color: blue to red (NES low to high).
(G) IHC staining of indicated proteins in human aorta tissues from young to old (n = 25). Spearman’s rank correlation was applied. Each point: one individual. Scale
bars: 50 μm.
(H) Boxplots of translational noise increase in tissue-enriched proteins during aging. Metrics: log2(1 + SD) and log2(1 + CV). SD, CV. Wilcoxon rank-sum test
applied.
(I) Left: density plots of GSVA scores using male-biased proteins in young groups across tissues (colors indicate sex groups). Right: density plots of GSVA scores
using female-biased proteins in young groups across tissues (colors indicate sex groups).
ll
Article
Figure S3. Age-related protein profile changes across tissues, related to Figure 2
(A) Top: pie charts of upregulated DEP subcellular location proportions. Top 2 Spearman’s correlation proteins are noted on the right. Bottom: abundance and
linear relationship with age for indicated proteins. Colors: tissues. Each point: one individual (Table S1).
(B) Top: pie charts of downregulated DEP subcellular location proportions. Bottom 2 Spearman’s correlation proteins are noted on the right. Bottom: abundance
and linear relationship with age for indicated proteins. Colors: tissues from (A). Each point: one individual (Table S1).
(C) Left: boxplots of Spearman’s correlation across tissues and aging upregulated DEP modules. Right: LOESS fitting trajectories of representative proteins in
each module. Significant tissues highlighted with colors from (A).
(D) Left: boxplots of Spearman’s correlation across tissues and aging downregulated DEP modules. Right: LOESS fitting trajectories of representative proteins in
each module. Significant tissues highlighted with colors from (A).
(E) Network diagram of GO terms enriched in upregulated DEP modules. Node pie charts: protein proportions. Pie sizes: enriched protein count; pie colors:
modules.
(F) Network diagram of GO terms enriched in downregulated DEP modules. Node pie charts: protein proportions. Pie sizes: enriched protein count; pie colors:
modules.
(G) Schematic of treating young hAECs with SAP protein.
(H) SA-β-Gal staining in hAECs treated with vehicle (n = 6) or SAP (n = 6). Scale bars, 100 μm.
(I) Tube formation analysis was performed in hAECs treated with vehicle (n = 6) or SAP (n = 6). Scale bars, 200 μm.
(J) Adhesion of monocytes assay was performed in hAECs treated with vehicle (n = 6) or SAP (n = 6). Scale bars, 100 μm.
(K) ELISA analysis of IL-6 was performed in hAECs treated with vehicle (n = 6) or SAP (n = 6).
Data are presented as mean ± SEM. Statistical significance between two groups was assessed using an unpaired two-tailed t test (H–K).
ll
Article
Figure S4. Protein hallmarks linked to human aging and aging-related diseases, related to Figure 3
(A) Immunostaining of p21Cip1, H3K9me3, IL-6, and collagen IV in various tissues (n = 22–62). Spearman’s correlation for coefficients and p values. Scale bars:
50 μm. Each point: one individual.
(B) Heatmap of downregulated epigenetic factor profile across tissues. Top 30 shared factors shown on right. Color key: blue to red (correlation coefficients low to
high).
(C) IHC staining of YTHDC1 in human liver (n = 50) and adrenal (n = 62) tissues from young to old. Spearman’s correlation for coefficients and p values. Each point:
one individual. Scale bars: 50 μm.
(D) Left: bar plot of complexes identified in each tissue. Right: pie charts of complex proportions in different functional groups across tissues.
(E) Left: dot plot of GSEA for aging-related chronic diseases. Dot sizes: − log10(p value); color: NES (light red to dark red). Right: network diagram of proteins
associated with aging-related chronic diseases. Node sizes: number of diseases; color: mean Spearman’s correlation coefficient (white to red).
ll
Article
Figure S5. Dynamic changes in proteomic clocks across tissues, related to Figure 4
(A) Bar plot of permutation importance proteins (importance > 1) (left) and top 5 importance proteins for each tissue aging clock (right).
(B) Heatmap of important proteins present in ≥4 tissue aging clocks. Color: permutation importance score (gray to red: low to high).
(C) Forest plot of TIMP3 importance across tissue clocks. Filled dots: importance >1; hollow points: importance ≤1; error bars: 90% confidence intervals.
(D) IHC staining of TIMP3 in spleen (n = 34), lymph (n = 25), adrenal (n = 62), pancreas (n = 34), and heart (n = 27). Each point: one individual. Spearman’s correlation
for coefficients and p values. Scale bars, 50 μm.
(E) We identified age-group-specific DEPs for early, middle, and late stages of aging. The percentage of these DEPs among detected proteins in each tissue is
shown (see STAR Methods).
(F) Bar graph of overlap between age-group-specific DEPs and lifespan-wide DEPs. Red: upregulated overlap; blue: downregulated overlap.
(G) Venn diagrams of overlap between age-group-specific and lifespan-wide DEPs. Left: upregulated overlap. Right: downregulated overlap. Circles and outer
box: different age stages.
(H) Enrichment analysis of DEPs overlapping between age-group-specific and lifespan-wide DEPs. Left: upregulated overlap. Right: downregulated overlap. Pie
chart sizes: protein count. Pie chart colors: proportion in different tissues.
(I) Relative number of time-resolved DEPs across tissues, normalized by the maximum in each tissue (see STAR Methods).
(J) Snapshots of time-resolved DEPs (normalized to detected proteins per tissue) at ages 30, 45, and 55 years. Colors: different tissues.
(K) Functional enrichment analysis of time-resolved DEPs at ages 30, 45, and 55 years. Pie charts: GO terms; pie size: enriched protein count; color: tissue
contribution.
ll
Article
Figure S6. Aging-related secretome dynamics and inter-organ communication, related to Figure 4
(A) Schematic of secretome categories. Secreted proteins are divided into 4 major groups and 21 subgroups based on biological functions.
(B) Pie chart of secreted protein proportions across categories. Number of proteins in each category is noted beside the name.
(C) Left: expression patterns of secreted proteins across tissues during aging (5 modules). Colors: mean Spearman’s correlation coefficient (blue to red). Middle:
boxplots of Spearman’s correlation across tissues and modules. Category proportions (colors from B) annotated on right. Right: representative GO terms en-
riched in each module. Dot sizes: protein counts; dot colors: − log10(p value) (white to black).
(D) Heatmap of SASP factors across tissues. Number of SASP factors per tissue on top; factors shared in ≥5 tissues on right. Colors: mean Spearman’s cor-
relation coefficient (white to red).
(E) Network diagram of tissue interactions during aging. Line thickness and color (white to red): interaction strength (low to high).
(F) Heatmap of ligand-receptor pairs across tissues during aging. Top: upregulated pairs (white to red: mean expression low to high). Bottom: downregulated
pairs (blue to white: mean expression low to high).
(G) Dot plot of interaction protein pairs and associated diseases. Left: enriched diseases from tissue pairs during aging (color and dot size: − log10(p value) low to
high). Right: interacting proteins of tissue pairs (color: interaction difference during aging.
(H) Ridge plot of GSEA for aging-related diseases. x axis: age correlation (Spearman’s correlation coefficient) of genes in pathways. Color: NES.
ll
Article
Figure S7. Plasma protein proxy aging clocks for different organs, related to Figure 6
(A) Bar plot of proteins with similar age-related trends in tissue and plasma (plasma-tissue paired DEPs). y axis: number of paired DEPs (below zero: commonly
downregulated; above zero: upregulated). x axis: different tissues. Gray bars: proteins with the same trend as plasma in a specific tissue; yellow bars: proteins
with similar trends in ≥2 tissues.
(B) Dot plot of plasma-tissue paired DEPs commonly downregulated with aging (Spearman’s rank correlation coefficient < 0, p < 0.05). Proteins differentially
expressed in ≥5 tissues are shown. x axis: proteins; y axis: tissues. Dot color: correlation of age-related change. Line plot above shows average plasma
abundance.
(C) ELISA analysis of HARS1 expression in plasma with age. Pearson’s correlation for coefficients and p values (n = 55, randomly selected individuals).
(D) Schematic of predicting tissue age using plasma proteins. Plasma and tissue proteomics data were integrated, and common proteins were selected. Elastic
net regression model built with tissue age as target. Plasma surrogate tissue clock developed after optimization and feature selection (see STAR Methods).
(E) Scatter plot of chronological age vs. predicted age (tissue age and plasma protein proxy tissue age). Correlation coefficient, p value, and MAE calculated using
Spearman’s correlation. Right bar chart: the top 10 proteins ranked by descending feature importance (see STAR Methods) in the plasma protein proxy clock.
(F) Dot plot of the Spearman’s correlation between plasma protein proxy aging clock predicted age and tissue age. x axis: predicted age by plasma protein proxy
aging clock using plasma protein abundance. y axis: tissue age predicted by tissue proteomic aging clocks using tissue protein abundance. Colors: white to red
(correlation coefficients low to high). Size: − log10(p value).
(G) Bar chart of feature counts for each plasma protein proxy aging clock.
(H) Dot plot of correlation between protein abundance (used in plasma protein proxy aging clock) and age in tissue, plasma, and their importance in the clock.
Spearman’s correlation for coefficients and p values. Dot color: correlation coefficient; dot size: − log10(p value).
ll
Article
Figure S8. Plasma senoproteins promote vascular endothelial cell senescence, related to Figure 7
(A–D) SA-β-Gal staining (A), western blot (B), migration assay (C), and tube formation (D) in hAECs treated with vehicle (n = 6 for A and C–E; n = 3 for B) or NOTCH3
(n = 6 for A and C–E; n = 3 for B). Scale bars, 100 μm.
(E) Dot plots of ELISA for IL-6 in hAECs treated with vehicle or recombinant proteins. Dot color: IL-6 concentration (fold induction, normalized by vehicle). Dot size:
p value (one-way ANOVA), n = 6.
(F) Top: bar plot of DEGs between recombinant protein groups (COMP, NOTCH3, HTRA1, SLPI, IGFBP7, GPNMB, and NEGR1) and the vehicle group. Upre-
gulated DEGs: red; downregulated DEGs: blue. Bottom: heatmap of relative GSVA scores for senescence-associated pathways. Colors: white to red (GSVA
scores low to high).
(G–I) SA-β-Gal (G), H3K9me3 (H), and LAP2 (I) staining in hVSMCs treated with vehicle or GPNMB. n = 6 (G), n = 300 nuclei (H and I). Scale bars: 200 μm (G), 20 μm
(H and I).
(J–L) SA-β-Gal (J), H3K9me3 (K), and LAP2 (L) staining in hVSMCs treated with vehicle or HTRA1. n = 6 (J), n = 300 nuclei (K and L). Scale bars: 200 μm (J), 20 μm (K
and L).
(M–O) SA-β-Gal (M), H3K9me3 (N), and LAP2 (O) staining in hVSMCs treated with vehicle or IGFBP7. n = 6 (M), n = 300 nuclei (N and O). Scale bars: 200 μm (M),
20 μm (N and O).
(P) Dot plots of ELISA for IL-6 in hVSMCs treated with GPNMB, HTRA1, or IGFBP7. Dot color: IL-6 concentration (fold induction, normalized by vehicle). Dot size:
p value (one-way ANOVA), n = 6.
Data are presented as mean ± SEM. Statistical significance between two groups was assessed using an unpaired two-tailed t test or Mann-Whitney test (A–E and
G–O).