Factores Genéticos Asociados
Factores Genéticos Asociados
Departamento de Medicina
Laboratorio de Investigación 10
Tesis Doctoral
María de los Ángeles Suárez Gestal
2010
El Dr. Antonio González Martínez-Pedrayo, investigador principal del Laboratorio de
Investigación 10 del Instituto de Investigación Sanitaria del Complejo Hospitalario
Universitario de Santiago de Compostela, y el Dr. Juan Jesús Gómez-Reino Carnota,
Jefe del Servicio de Reumatología del Hospital Clínico Universitario de Santiago de
Compostela y Profesor Titular del Departamento de Medicina y Catedrático de la
Universidad de Santiago de Compostela,
CERTIFICAN QUE:
El presente trabajo que lleva por título: Factores genéticos asociados con
predisposición a Lupus Eritematoso Sistémico: Confirmación, efecto funcional,
repercusión fenotípica y relación con la susceptibilidad a la Artritis Reumatoide,
realizado por María de los Ángeles Suárez Gestal en el Departamento de Medicina de la
Universidad de Santiago de Compostela bajo nuestra dirección, ha sido revisado y está
en disposición de ser presentado para optar al grado de Doctora en Biología.
Fdo: Dr. Antonio González Martínez-Pedrayo Fdo: Dr. Juan J. Gómez-Reino Carnota
A mi hermana Merchi
Agradecimientos
El camino ha sido duro y largo, con unos momentos mejores que otros,
pero todo este esfuerzo ha merecido la pena, no sólo por los objetivos
alcanzados en mi trabajo, sino también porque durante estos años he conocido a
gente maravillosa que, junto a mi familia y amigos me han apoyado desde el
principio hasta el final de mi tesis.
Y siguiendo con más gente de Pontevedra, le quiero dar las gracias a Rita,
porque en poco tiempo se ha convertido en una amiga en la que se puede
confiar. A Peon y Claudio, por el buen rato que hemos pasado en las pachangas,
en el rafting, en la carrera de orientación o tomando unas cañas. Y a Toño,
porque durante un tiempo ha sido un gran apoyo para mí, hablar con él me
tranquilizaba y me daba fuerzas para seguir. Por el cariño que me transmitió,
porque los buenos momentos fueron muchos y esos son los que quedan en el
recuerdo y porque sigo disfrutando de su compañía.
Índice
Abreviaturas 1
Introducción 7
1. Lupus Eritematoso Sistémico 9
1.1. Patogenia del LES 10
1.2. Clínica del LES 14
1.3. El LES como enfermedad compleja 16
2. Influencia del componente genético en la variabilidad clínica del
LES 17
3. Estudio de la genética del LES 19
3.1. Estudios de ligamiento 19
3.2. Estudios de asociación 21
3.2.1. PDCD1 23
3.3. Estudios de asociación de genoma completo 27
3.3.1. GWAS en LES 30
3.4. Factores genéticos de LES confirmados 37
3.4.1. HLA 37
3.4.2. Factores del complemento 40
3.4.3. FCGRs 42
3.4.4. TREX1 43
3.4.5. IRF5 45
3.4.6. PTPN22 46
3.4.7. STAT4 47
3.4.8. ITGAM 49
3.4.9. C8orf13-BLK 50
3.4.10. BANK1 51
Factores genéticos asociados con predisposición a LES
Abreviaturas
1
Factores genéticos asociados con predisposición a LES
2
Abreviaturas
3
Factores genéticos asociados con predisposición a LES
4
Abreviaturas
5
INTRODUCCIÓN
Introducción
Introducción
9
Factores genéticos asociados con predisposición a LES
El LES es una enfermedad que afecta con una frecuencia 9 veces mayor a
mujeres que a hombres, sobre todo a mujeres en edad fértil. La prevalencia en
población europea es del 0.01-0.06 %, mientras que es tres o cuatro veces más
frecuente en poblaciones asiática, africana e hispano-americana (Petri M y col.,
2002). Esta heterogeneidad étnica junto con la heterogeneidad genética pueden
contribuir a la complejidad de sus manifestaciones clínicas.
10
Introducción
11
Factores genéticos asociados con predisposición a LES
“asesinas naturales”. Estos dos tipos de células serían las encargadas de limitar
la expansión de las células Th y B autorreactivas, por lo que la alteración de su
función produce un defecto en la tolerancia periférica.
12
Introducción
13
Factores genéticos asociados con predisposición a LES
expresión del factor asociado al receptor del factor de necrosis tumoral (TRAF)
6, de la quinasa 1 asociada al receptor de IL1 (IRAK1) (Taganov KD y col.,
2006), del transductor de señal y activador de la transcripción (STAT) 4 y del
factor regulador de IFN (IRF) 5 (Tang Y y col., 2009). En concordancia con
estas funciones, miR-146 tiene una expresión disminuida en pacientes con LES,
correlacionando negativamente con la actividad de la enfermedad y los niveles
de IFN (Tang Y y col., 2009).
14
Introducción
Tabla 1. Criterios de clasificación del LES establecidos en 1982 (Tan EM y col., 1982)
y actualizados en 1997 (Hochberg MC y col., 1997).
15
Factores genéticos asociados con predisposición a LES
16
Introducción
17
Factores genéticos asociados con predisposición a LES
18
Introducción
Los dos tipos de estudios que se utilizan para buscar factores genéticos de
susceptibilidad son los estudios de ligamiento y los estudios de asociación.
19
Factores genéticos asociados con predisposición a LES
raras con grandes efectos, es decir, que provocan una alta probabilidad de
desarrollar la enfermedad. Sin embargo, su aplicación a los fenotipos complejos,
como el LES, es más problemática porque no existe un modelo que explique
adecuadamente el patrón de herencia y porque los factores genéticos de
susceptibilidad para estas enfermedades suelen ser muchos y tener un efecto
pequeño. De hecho, los estudios de ligamiento de genoma completo realizados
hasta el momento en diversas poblaciones (europea, hispana, afro-americana y
asiática) indican que la susceptibilidad al LES vendría dada por múltiples loci
de pequeño efecto y de magnitud similar (Cantor RM y col., 2004; Gaffney PM
y col., 1998; Gaffney PM y col., 2000; Gray-McGuire C y col., 2000; Johansson
CM y col., 2004; Koskenmies S y col., 2004; Lindqvist AK y col., 2000; Moser
KL y col., 1998; Nath SK y col., 2004; Shai R y col., 1999, Rao S y col., 2001;
Olson JM y col., 2002). Sin embargo, no hay mucha concordancia entre los
resultados. De los casi 60 loci que han demostrado evidencias de ligamiento con
LES en estudios de genoma completo, sólo 17 han sido reproducidos en, al
menos, dos estudios independientes: 1p36, 1q23-24, 2q23-33, 2q37, 4p15-13,
4q32, 5p15, 6p22-q15, 11q23, 12q24, 14q23, 16p13, 16q12-13, 17q11, 18q21,
19q13.1 y 20p13. En consecuencia, para encontrar alguna evidencia consistente
en los estudios de ligamiento realizados, se han llevado a cabo dos meta-análisis
(Lee YH y col., 2005; Forabosco P y col., 2006). Ambos confirmaron los loci
6p22-q15, 16p12-q12, 20p12-q13 y 2q31-q34, proponiéndose como genes
causantes del ligamiento de los dos primeros loci al MHC y a CARD15/NOD2
(caspase recruitment domain 15 / nucleotide-binding oligomerization domain
containing 2), respectivamente. De todos modos, el MHC no tiene un efecto tan
fuerte como en otras enfermedades autoinmunes como la artritis reumatoide
(AR).
20
Introducción
21
Factores genéticos asociados con predisposición a LES
22
Introducción
3.2.1. PDCD1
23
Factores genéticos asociados con predisposición a LES
24
Introducción
25
Factores genéticos asociados con predisposición a LES
26
Introducción
27
Factores genéticos asociados con predisposición a LES
Tras genotipados a gran escala como los que se realizan con estos chips,
es necesario llevar a cabo un control de calidad a tres niveles: SNPs, muestras y
genotipos. En cuanto a los SNPs, se comprueba la eficiencia de genotipado y si
la frecuencia del alelo menor se encuentra en equilibrio de Hardy-Weinberg así
como si está en concordancia con los datos de HapMap y con los duplicados
internos. Con respecto a las muestras, se comprueba también la eficiencia de
genotipado y la concordancia entre el sexo recogido y el observado. Y con los
datos de genotipo se comprueba si existe algún problema de subestructura
poblacional y si hay muestras que puedan tener una relación de parentesco
oculta, lo que puede dar lugar a falsos positivos.
28
Introducción
29
Factores genéticos asociados con predisposición a LES
30
Introducción
8.49x10-11
col., 2008
31
Factores genéticos asociados con predisposición a LES
Tabla 2. Resumen de los hallazgos principales de los GWAS en LES. Para cada
estudio se detallan los SNPs asociados, el locus en el que se encuentran, la posición en el
cromosoma, los alelos, si la asociación alcanzó el nivel de significación requerido para los GWAS
y el valor de P en el análisis conjunto de todos los resultados obtenidos en el mismo estudio. a En
negrita, el alelo de riesgo.
Un segundo estudio analizó 502033 SNPs en 1311 casos con LES y 3340
controles de población europeo-americana, realizando posteriormente una fase
de confirmación de los resultados más destacables en 793 casos con LES y 857
controles de población sueca (Hom G y col., 2008). Sus resultados mostraron
también la asociación significativa de polimorfismos en la región cromosómica
que contiene el gen ITGAM. Además, vieron asociación de un locus que hasta
ahora no había sido relacionado con LES: la región promotora de los genes de la
quinasa de linfocitos B (BLK) y de C8orf13, que comparten la misma región
promotora aunque se transcriben en dirección opuesta (Hom G y col., 2008)
(Figura 3).
32
Introducción
33
Factores genéticos asociados con predisposición a LES
34
Introducción
35
Factores genéticos asociados con predisposición a LES
36
Introducción
3.4.1. HLA
Los primeros genes que fueron descubiertos como factores de riesgo para
el LES fueron los que codifican para el HLA. Las moléculas del HLA están
37
Factores genéticos asociados con predisposición a LES
El estudio que más contribuyó a definir la asociación del MHC con LES
incluyó unos 50 microsatélites analizados en 334 familias multicaso. En este
análisis quedó clara la importancia de los haplotipos que incluyen HLA-DRB1 y
DQB1, concretamente los correspondientes al HLA-DR2 (DRB1*1501-
DQB1*0602), HLA-DR3 (DRB1*0301-DQB1*0201) y, con una menor
importancia, al HLA-DR8 (DRB1*0801-DQB1*0402) (Graham RR y col.,
2002). Este estudio delimitó una región de 1 Mb, que engloba la región del
MHC de clase II y III, pero no fue posible precisar más debido al alto LD de la
zona. Los haplotipos portadores del HLA-DR3 confieren mayor riesgo que los
portadores del HLA-DR2 y los individuos homocigotos para DR3 o
heterocigotos DR3/DR2 muestran el riesgo más alto. Además, estos haplotipos
se han asociado con la producción de autoanticuerpos característicos del LES
(Graham RR y col., 2007b; Sebastiani GD y col., 2009). Concretamente, el
haplotipo DR2 se asocia con la producción de anti-Sm y anti-ADN de doble
cadena, mientras que DR3 está relacionado con mayores niveles de anti-Ro y
anti-La. Los GWAS publicados recientemente (SLEGEN y col., 2008; Hom G y
col., 2008; Graham RR y col., 2008) han confirmado que los marcadores
localizados en la región del HLA de clase II son los más fuertemente asociados
con la enfermedad, aunque su contribución al LES no es mucho mayor que la de
otros factores genéticos encontrados (Figura 6), por lo que el efecto del HLA
no es comparable con el observado en otras enfermedades autoinmunes como
38
Introducción
39
Factores genéticos asociados con predisposición a LES
11
10
6
OR
K
A, q
ST 5
FC AM
TN 22
AA 4
FC IP3
B
C2
IR 4
1
BA 1
TN A
IT B
3A
PT 2A
K
42
P2
FC K1
F
AK
KI S F
C4 C1
AT
or 25.
BL
PX
3
C4
HL
IR
PN
15
R
EC
N
G
FA
3-
G
C8 1q
f1
M
Figura 6. Comparación del efecto de los genes asociados al LES. Como medida del
tamaño del efecto se usaron las ORs (Moser KL y col., 2009).
40
Introducción
41
Factores genéticos asociados con predisposición a LES
3.4.3. FCGRs
42
Introducción
3.4.4. TREX1
43
Factores genéticos asociados con predisposición a LES
44
Introducción
gen un buen candidato para tener un papel en dicha respuesta característica del
LES.
3.4.5. IRF5
45
Factores genéticos asociados con predisposición a LES
3.4.6. PTPN22
46
Introducción
3.4.7. STAT4
47
Factores genéticos asociados con predisposición a LES
48
Introducción
3.4.8. ITGAM
ITGAM codifica para la integrina α-M que dimeriza con la integrina β-2
(ITGB2 o CD18), formando el receptor del complemento 3 (también conocido
como CD11b o Mac-1). Este complejo se expresa en la superficie celular de
macrófagos, monocitos, DCs y neutrófilos. Las funciones de ITGAM son muy
variadas e incluyen su participación en la adhesión y transmigración de los
leucocitos a través del endotelio (Dunne JL y col., 2003), la activación de
49
Factores genéticos asociados con predisposición a LES
3.4.9. C8orf13-BLK
50
Introducción
3.4.10. BANK1
51
Factores genéticos asociados con predisposición a LES
52
Introducción
3.4.12. KIAA1542-IRF7
3.4.13. PXK
53
Factores genéticos asociados con predisposición a LES
54
Introducción
El gen TNFAIP3 y la región intergénica 6q23 son dos loci vecinos que se
encuentran en bloques haplotípicos distintos y que están asociados a varias
enfermedades autoinmunes incluyendo AR (Wellcome Trust Case Control
Consortium, 2007; Thomson W y col., 2007; Dieguez-Gonzalez R y col., 2009)
y LES (Graham RR y col., 2008; Musone SL y col., 2008). Sin embargo, los
detalles de la asociación no coinciden. El SNP de riesgo que mostraba una
55
Factores genéticos asociados con predisposición a LES
56
Introducción
3.4.16. TNIP1
57
Factores genéticos asociados con predisposición a LES
3.4.17. PRDM1-ATG5
3.4.18. JAZF1
La región promotora del gen JAZF1 mostró asociación con LES, con P <
5 x 10-8 (Gateva V y col., 2009). La misma variante de susceptibilidad
(rs849142) se observó previamente en diabetes tipo 2 (T2D) (Zeggini E y col.,
2008). Otro SNP independiente (rs10486567), cercano al gen, también mostró
asociación con susceptibilidad al cáncer de próstata (Thomas G y col., 2008),
58
Introducción
pero no se asoció a LES (Gateva V y col., 2009). JAZF1 codifica para un factor
de transcripción que regula la expresión de numerosos genes, por lo que se
desconoce cuál podría ser su función específica en el LES.
3.4.19. UHRF1BP1
3.4.20. IL10
59
Factores genéticos asociados con predisposición a LES
Hay muchos genes que han sido asociados con LES en algún estudio pero
que no han sido confirmados con posterioridad. Aquí sólo se tratarán dos que
por la solidez de los estudios que los apoyan pueden ser genuinos factores de
riesgo aunque no hayan sido todavía replicados de forma definitiva.
3.5.1. TYK2
60
Introducción
3.5.2. LY9
61
Factores genéticos asociados con predisposición a LES
62
Introducción
63
Factores genéticos asociados con predisposición a LES
64
Introducción
65
Factores genéticos asociados con predisposición a LES
Los GWAS han sido una potente herramienta para la investigación del
componente genético de las enfermedades complejas, identificando gran
cantidad de variantes genéticas de susceptibilidad. La mayoría de asociaciones
encontradas hasta ahora muestran ORs ≤ 1.5 (Figura 6). Este pequeño efecto y
el gran número de genes de susceptibilidad que se están encontrando indica que
en el LES participan múltiples factores genéticos ya que cada uno de ellos
confiere un pequeño aumento en el riesgo a padecer la enfermedad. Sin
embargo, a pesar de ser numerosos explican sólo una parte pequeña de la
heredabilidad (proporción del fenotipo atribuible a factores genéticos). Así, en
el estudio GWA del SLEGEN se estimó que PXK, HLA, IRF5, KIAA1542 e
ITGAM explican tan sólo un 15 % de la heredabilidad (SLEGEN y col., 2008).
Estos resultados llevan a preguntarse dónde puede residir la heredabilidad
todavía no identificada.
Varias han sido las sugerencias sobre las distintas razones que podrían
explicar la falta de detección de una proporción considerable de la heredabilidad.
En primer lugar, es posible que aún no se hayan identificado muchas de las
variantes comunes de pequeño efecto. Otra posibilidad es que las variantes
causales por descubrir sean variantes de baja frecuencia (1-5 %) o variantes
raras (frecuencia < 0.5 %), que no están bien cubiertas por los GWAS actuales.
Además, es posible que estas variantes poco frecuentes tengan un efecto más
notable. Sin embargo, su efecto no puede ser muy fuerte ya que no han sido
detectadas por los estudios de ligamiento (Manolio TA y col., 2009). Un
ejemplo de variantes de este tipo son las variantes raras en TREX1 (Lee-Kirsch
MA y col., 2007) que ya han sido comentadas. Otra posible fuente de
heredabilidad no explicada son las variantes estructurales, como las CNVs.
66
Introducción
67
Factores genéticos asociados con predisposición a LES
Como parte del estudio GWA llevado a cabo por el consorcio SLEGEN,
se exploró si había evidencias de epistasis entre los genes asociados, pero no las
encontraron (SLEGEN y col., 2008). Otros estudios analizaron la interacción
entre genes que forman parte de la misma ruta funcional. Así, STAT4 e IRF5
son miembros de la ruta de IFN de tipo I, por lo que sus polimorfismos de
susceptibilidad serían candidatos a interaccionar. Lo que se observó en un
estudio de 500 pacientes y 500 controles suecos fue que con cada alelo de riesgo
del rs7582694 de STAT4 (que define el haplotipo de riesgo y tiene r2 = 1 con el
rs7574865) y de dos SNPs (indel del promotor y rs10488631) de IRF5 la OR se
incrementaba en 1.82 (Sigurdsson S y col., 2008b). Un efecto incrementado al
combinar estos dos factores genéticos se describió en otro estudio de LES con
muestras europeas e hispanas (Abelson AK y col., 2009), así como en pacientes
con síndrome de Sjögren (SS) (Nordmark G y col., 2009) y en pacientes con
esclerosis sistémica cutánea difusa (Dieudé P y col., 2009). Sin embargo, los
aumentos de riesgo observados en estos estudios no han detectado desviación de
la suma de susceptibilidad que confiere cada alelo por sí mismo, por lo que no
hay evidencias de interacción epistática entre estos dos factores.
Otro factor que participa en la ruta del IFN es TYK2, por lo que también
se ha hipotetizado su posible interacción con IRF5. De hecho, un estudio
encontró epistasis significativa (P = 0.014) entre los SNPs rs10954213 de IRF5
y rs2304256 de TYK2. La combinación genotípica que más contribuyó a la
interacción era la de los homocigotos para los alelos de riesgo de ambos SNPs
(OR = 2.73) (Hellquist A y col., 2009). Aún así, el tamaño muestral de este
estudio no es muy grande (300 pacientes y 300 controles), por lo que sería
necesaria la confirmación de este resultado por estudios independientes de
mayor tamaño.
68
Introducción
69
Factores genéticos asociados con predisposición a LES
70
Introducción
71
Factores genéticos asociados con predisposición a LES
72
Introducción
algunas de estas señales no están bien recogidas en nuestro estudio previo. Las
más interesantes son las asociaciones encontradas en LES debido a su fuerte
efecto y a que una se corresponde con un nsSNP que podría tener un papel
causal en la susceptibilidad a la enfermedad.
73
HIPÓTESIS
Hipótesis
Hipótesis
77
OBJETIVOS
Objetivos
Objetivos
81
PROCEDIMIENTO EXPERIMENTAL Y
RESULTADOS
Procedimiento experimental y resultados
Esquema general
Publicación 1:
Publicación 2:
85
Factores genéticos asociados con predisposición a LES
Publicación 3:
Publicación 4:
Publicación 5:
86
Procedimiento experimental y resultados
Publicación 6:
Publicación 7:
87
1. Factores genéticos de LES
Procedimiento experimental y resultados
1Laboratorio de Investigacion 10 and Rheumatology Unit, Hospital Clinico Universitario de Santiago, Santiago de Compostela 15706, Spain
2Paediatrics Department, Albert Szent-Györgyi Medical and Pharmaceutical Centre, University of Szeged, Szeged 6721, Hungary
3Instituteof Clinical Biochemistry, Martin Faculty Hospital, Jessenius Medical Faculty, Kollárova 2, 036 59 Martin, Slovakia
4Internal Medicine, Research Laboratory in Autoimmune Diseases, Hospital Vall d'Hebron, 08035 Barcelona, Spain
5Ospedale S Camillo-Forlanini, U O Complessa di Reumatologia, 00151 Roma, Italy
6Molecular Biology and Immunogenetics Department, Institute of Rheumatology, 128 50 Prague 2, Czech Republic
7Rheumatology Department, Hospital Garcia de Orta, Almada, Portugal
8Rheumatology Research Unit, Instituto Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Portugal
9Department of Histocompatibility and Immunology, Evangelismos Hospital, 10676 Athens, Greece
10Clinical Immunology, University of Milan and Fondazione IRCCS Ospedale Maggiore Policlinico, Mangiagalli e Regina Elena, 20122 Milan, Italy
11Pathophysiology Department, Athens University Medical School, Athens 115 27, Greece
12Department of Functional Biology, Hospital Universitario Central de Asturias, Universidad de Oviedo, Oviedo 33006, Spain
13INIBIC-CH Universitario A Coruña, 15006 A Coruña, Spain
14Dept Medical Sciences and IRCAD, Eastern Piedmont University, 28100 Novara, Italy
15Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, 9713 Groningen, The Netherlands
16Rheumatology Unit Hospital 12 de Octubre, 28041 Madrid, Spain
17Division of Clinical Immunology, Department of Internal Medicine of the Hannover Medical School, D-30625 Hannover, Germany
18Rheumatology Unit, Second University of Naples, 81100 Naples, Italy
19Department of Medicine, University of Santiago de Compostela, Santiago de Compostela, 15706, Spain
Received: 24 Mar 2009 Revisions requested: 29 Apr 2009 Revisions received: 8 May 2009 Accepted: 14 May 2009 Published: 14 May 2009
Abstract
Introduction We aimed to replicate association of newly 10-5), as well as association with the X chromosome MECP2
identified systemic lupus erythematosus (SLE) loci. gene (OR = 1.26, P = 0.00085 in women), which had only been
reported in a single study, and association with four other loci,
Methods We selected the most associated SNP in 10 SLE loci. 1q25.1 (OR = 0.81, P = 0.0001), PXK (OR = 1.19, P =
These 10 SNPs were analysed in 1,579 patients with SLE and 0.0038), BANK1 (OR = 0.83, P = 0.006) and KIAA1542 (OR
1,726 controls of European origin by single-base extension. = 0.84, P = 0.001), which have been identified in a genome-
Comparison of allele frequencies between cases and controls wide association study, but not found in any other study. All
was done with the Mantel–Haenszel approach to account for these replications showed the same disease-associated allele
heterogeneity between sample collections. as originally reported. No association was found with the LY9
SNP, which had been reported in a single study.
Results A previously controversial association with a SNP in the Conclusions Our results confirm nine SLE loci. For six of them,
TYK2 gene was replicated (odds ratio (OR) = 0.79, P = 2.5 × TYK2, MECP2, 1q25.1, PXK, BANK1 and KIAA1542, this
BANK1: B-cell scaffold protein with ankyrin repeats 1; BLK: B-lymphoid tyrosine kinase; GWA: genome-wide association; IL: interleukin; ITGAM:
integrin alpha M; LY9: lymphocyte antigen 9; MECP2: methyl CpG binding protein 2; OR: odds ratio; PXK: PX domain containing serine/threonine
kinase; SLE: systemic lupus erythematosus; SLEGEN: International Consortium for Systemic Lupus Erythematosus Genetics; SNP: single nucleotide
polymorphism; STAT4: signal transducer and activator of transcription; TYK2: tyrosine kinase 2.
Page 1 of 9
(page number not for citation purposes)
91
Factores genéticos asociados con predisposición a LES
replication is important. The other three loci, ITGAM, STAT4 and bias of SLE, contrary to what has been proposed. In addition,
C8orf13-BLK, were already clearly confirmed. Our results also none of the other associations seems important in this respect.
suggest that MECP2 association has no influence in the sex
Introduction pean countries (see Table S1 in Additional data file 1). Most of
Systemic lupus erythematosus (SLE) is a complex autoim- these samples have already been described [17]. Two new
mune disease of wide variability in its manifestations and clini- sample collections were from Asturias, Spain and Almada,
cal evolution that characteristically involves multiple Portugal. Each recruiting centre was asked for about 100 SLE
autoantibodies against ubiquitous nuclear antigens. Its patients and 100 ethnically matched controls. A total of 1,579
genetic component is very significant, as shown by a sibling cases and 1,726 controls were obtained in this way. All SLE
recurrence risk ratio of 20 and a 10-fold excess in SLE con- patients met the revised American College of Rheumatology
cordance between monozygotic twins over dizygotic twins classification criteria [18]. Clinical characteristics of the
[1,2]. patients are provided in Table S2 in Additional data file 1.
Patients and controls gave written informed consent. Sample
Linkage studies have indicated that this genetic component is collection was approved by the respective ethical committees.
due to multiple low-penetrance common genetic factors [1].
Only a few factors had consistently been demonstrated until Genotyping
2008: the class II HLA alleles, low-affinity receptors for the We selected a SNP for each of the 10 associated loci that we
constant fraction of IgG, and the PTPN22 and IRF5 genes. intended to replicate (Table 1). The SNPs were selected
This scenario has been dramatically improved by new technol- because they were strongly associated with SLE or because
ogies and genome resources [2]. Four genome-wide associa- they were described as probable causal polymorphisms.
tion (GWA) studies were published in 2008 [3-6] that, These 10 SNPs were amplified in a single PCR with the Qia-
together with other large-scale studies, have greatly enlarged gen Multiplex PCR kit (Qiagen, Chatsworth, CA, USA) with 20
the number of convincing SLE-associated loci. Not all of the ng genomic DNA and 0.2 PM of each primer (for primers and
newly described findings, however, have attained the same probes, see Table S3 in Additional data file 1). The PCR prod-
degree of confirmation [2]. Some of them are already defini- ucts were purified by digestion with Exonuclease I (Epicentre,
tively confirmed by replication in different sample collections Madison, WI, USA) and shrimp alkaline phosphatase (GE
by the same authors and also by independent authors in sep- Healthcare, Barcelona, Spain). Purified PCR products were
arate studies (Table 1). In this group are the SLE associations genotyped by single-base extension with the SNaPshot Multi-
with the ITGAM [3,4,6,7], STAT4 [3,4,6,8-12] and C8orf13- plex Kit (Applied Biosystems, Foster City, CA, USA) and spe-
BLK regions [3,4,6]. Other findings are very solid but they still cific probes. After a second purification with shrimp alkaline
require confirmation by independent studies. In this group are phosphatase (GE Healthcare), samples were analysed in the
the associated loci that were only reported in a single GWA Abi Prism 3130xl Genetic Analyzer (Applied Biosystems) and
study but not in the other studies, such as BANK1 [5], PXK genotypes assigned by the GeneMapper software. All geno-
[3], KIAA1542 [3] and 1q25.1 [3], or those that were reported type calls were manually reviewed and conflicting results were
in a single large study but not in any of the four GWA studies, liberally re-assayed or re-genotyped by sequencing with the
such as MECP2 [13] and LY9 [14]. Finally, the TYK2 associ- Big Dye Ready Reaction Kit v 3.1 (Applied Biosystems).
ation is more controversial because it was found in a large Sequence reactions followed the kit manufacturer protocol
study with Scandinavian families [15], partially replicated in a and were also analysed in the Abi Prism 3130xl Genetic Ana-
large study of UK families [16], and excluded in one of the lyzer.
GWA studies [3].
Statistical analysis
In the present paper, therefore, we have analysed SLE associ- Some of the sample collections in our study have already been
ation to each of these loci in more than 1,500 SLE patients used for the analysis of specific associations included in this
and 1,700 controls – and all of them except LY9 have been project. They have been excluded from the relevant analyses
clearly replicated. In addition, we have found that many of to avoid data duplication; this circumstance is detailed in Table
these loci are also important for SLE in men where data from S4 in Additional data file 1, where raw genotype data from
previous reports is almost completely absent. each sample collection are reported. Hardy-Weinberg equilib-
rium tests in control samples were performed with Haploview
Materials and methods with a threshold of 0.05 uncorrected for multiple tests [19].
Sample collection Other statistical analyses were carried out in a customized ver-
We used DNA samples from SLE patients and ethnically sion of the Statistica 7.0 program (StatSoft, Tulsa, OK, USA).
matched healthy controls of 16 collections from nine Euro-
Page 2 of 9
(page number not for citation purposes)
92
Procedimiento experimental y resultados
Table 1
Newly systemic lupus erythematosus-associated loci that were examined with previous evidence of association
Locus Chr. Location SNP Allelesa OR (95% CI) Associated Sample size Population Reference
ITGAM 16 Exon 3 rs1143679 G/A 1.78 (1.6 to 2.0) Yes 3,818 European American [7]
1.55 (1.2 to 2.0) Yes 1,289 African American [7]
2.07 (1.3 to 3.4) Yes 271 Gullah [7]
Other SNPs Yes [3,4,6]
STAT4 2 Intron 3 rs7574865 G/T 1.5 (1.2 to 1.8) Yes 3,057 European [3]
1.50 (1.4 to 1.7) Yes 4,651 European American [4]
1.55 (1.3 to 1.8) Yes 2,287 European American [8]
1.61 (1.4 to 1.9) Yes 2,495 Japanese [9]
1.62 (1.2 to 2.2) Yes 565 Colombians [10]
1.56 (1.4 to 1.7) Yes 3,958 European [12]
Other SNPs Yes [6,11]
C8orf13-BLK 8 Intergenic rs13277113 G/A 1.39 (1.3 to 1.5) Yes 6,301 European American [4]
Other SNPs Yes [3,6]
TYK2 19 Exon 8 rs2304256 C/A 0.625 (0.5 to 0.8) Yes 966 Swedish/Finnish [15]
105:127b No 380c European/Indo- [16]
Pakistani
Other SNPs No [3]
MECP2 X Intron 2 rs17435 A/T 1.58 (1.3 to 1.9) Yes 1,364 Korean [13]
1.29 (1.1 to 1.5) Yes 2,160 European [13]
1q25.1 1 Intergenic rs10798269 G/A 0.82 (0.8 to 0.9) Yes 6,728 European [3]
BANK1 4 Intron 1 rs17266594 T/C 0.74 (0.6 to 0.9) Yes 927 Scandinavian [5]
0.70 (0.5 to 0.9) Yes 576 German [5]
0.63 (0.5 to 0.8) Yes 450 Italian [5]
0.78 (0.6 to 0.9) Yes 1,136 Spanish [5]
0.58 (0.4 to 0.8) Yes 620 Argentinian [5]
KIAA1542 11 Intron 4 rs4963128 G/A 0.78 (0.7 to 0.9) Yes 6,728 European [3]
PXK 3 Intron 5 rs6445975 T/G 1.25 (1.2 to 1.4) Yes 6,728 European [3]
LY9 1 Exon 8 rs509749 A/G 377:403d Yes 510c UK Caucasian [14]
237:251d No 270c Canadian [14]
Loci ordered as presented in Table 2. The SNPs selected for replication are detailed. Chr, chromosome; CI, confidence interval; OR, odds ratio.
aMajor/minor alleles. bTransmitted:untransmitted. cNumber of families. dObserved:expected.
Comparison of cases and controls was carried out with the with the Breslow–Day test, is excluded. Significant heteroge-
Mantel–Haenszel approach because allele frequency differ- neity of effects is therefore excluded by the Breslow–Day test
ences are probable between sample collections even if spe- and allele frequency heterogeneity is accounted for with the
cific effects on the phenotype are constant. Spurious false Mantel–Haenszel approach. These analyses were also con-
positive or false negative results therefore become likely if the ducted after stratifying the samples by gender. Univariate
allele differences are not accounted for. To avoid this, the Man- logistic regression models were used to test the fit to the data
tel–Haenszel approach combines effect sizes taken as the of additive, recessive and dominant genetic models. Statistical
odds ratio (OR) in each stratum allowing for heterogeneity in power was estimated with the Power and sample size calcula-
allele frequencies. This approach provides an accurate com- tions software [20].
bined statistic if the heterogeneity of effect sizes, evaluated
Page 3 of 9
(page number not for citation purposes)
93
Factores genéticos asociados con predisposición a LES
Results with an effect size equivalent to OR > 1.15 with P < 0.05 (or
A total of 1,579 SLE patients and 1,726 controls from 16 OR > 1.23 for P < 0.001).
European collections were available for study (Tables S1 and
S2 in Additional data file 1). The genotyping call rate was In addition to these important results for replication, we found
99.9% and the genotypes in controls were in Hardy-Weinberg association with the three loci that have already been repli-
equilibrium. Individual collection data for each SNP is shown cated in GWA studies: rs1143679 in ITGAM (OR = 1.70, P
in Tables S4 and S5 in Additional data file 1. Combined anal- = 1.1 × 10-16), rs7574865 in STAT4 (OR = 1.62, P = 2.4 ×
ysis of the SNP effects across our sample collections was per- 10-12) and rs13277113 in C8orf13-BLK (OR = 1.34, P = 5.1
formed with the Mantel–Haenszel approach, which is a × 10-7). The effect sizes of these three association signals
method correcting for variability in allele frequencies between (that is, their ORs) were larger than for all the other signals,
collections provided that the effect sizes (that is, ORs) are not perhaps explaining the more consistent replication of their
significantly divergent. This condition was fulfilled because no association. Genotype comparisons for the different SNPs
significant heterogeneity in OR was detected for any of the were concordant with an additive genetic model and yielded
SNPs (Table 2, final column). very similar results to the allele frequency analyses (data not
shown).
The combined data showed significant differences between
SLE cases and controls for eight of the nine SNPs located in Combined analysis was also conducted in women (Table 3).
autosomal chromosomes (Table 2). All of the significant differ- This was particularly necessary for the MECP2 SNP rs17435,
ences between cases and controls were in the same direction located in the X chromosome. This SNP showed a significant
as originally reported (Tables 1 and 2). We found association difference between SLE women and control women and with
of the four SNPs that have been reported in a single GWA and the same disease-associated allele as previously reported (OR
not yet replicated by independent studies: rs10798269 in = 1.26, P = 0.00085). The SNPs placed in the autosomes
1q25.1 (OR = 0.81, P = 0.00013), rs6445975 in PXK (OR = showed similar results to those obtained in the unstratified
1.19, P = 0.0038), rs17266594 in BANK1 (OR = 0.83, P = analysis. There were only less significant P values due to the
0.0062) and rs4963128 in KIAA1542 (OR = 0.84, P = smaller sample size, but the effect sizes (expressed as ORs)
0.0011). There was also significant association of two of the remained largely unchanged. The BANK1 SNP was not asso-
three SNPs that were described in large studies but that were ciated in women, but this was the SNP with fewer available
not observed in any of the GWA studies: rs2304256 in TYK2 samples because we have excluded from this analysis the
(OR = 0.79, P = 2.5 × 10-5) and rs17435 in MECP2 (analysis sample collections that have previously been reported (power
of this SNP was performed separately in women and men was 0.68 for P = 0.05 and OR = 0.78, which was previously
because this gene is in chromosome X; see below). Only reported in Spanish samples) [5].
rs509749 in LY9 was similar in cases and controls. Our study
had sufficient power (80%) to detect association at this SNP No previous detailed information of men with SLE has been
published for any of these associated loci, although in a report
Table 2
Combined analysis of allele frequency differences between SLE cases and controls for nine autosomal loci
Minor allele frequency (%)a Mantel – Haenszel analysis Breslow – Day test
rs1143679 (ITGAM) 23.2 (730/3,152) 15.1 (521/3,448) 1.70 (1.5 to 1.9) 1.1 × 10-16 0.5
rs7574865 (STAT4) 32.8 (709/2,162) 23.2 (485/2,092) 1.62 (1.4 to 1.9) 2.4 × 10-12 1.0
rs13277113 (C8orf13-BLK) 30.9 (874/2,824) 25.7 (776/3,024) 1.34 (1.2 to 1.5) 5.1 × 10-7 1.0
rs2304256 (TYK2) 23.3 (733/3,152) 27.8 (960/3,450) 0.79 (0.7 to 0.9) 2.5 × 10-5 0.6
rs10798269 (1q25.1) 27.3 (861/3,158) 31.8 (1,098/3,452) 0.81 (0.7 to 0.9) 0.00013 0.2
rs17266594 (BANK1) 24 (526/2,192) 27.6 (624/2,260) 0.83 (0.7 to 0.9) 0.0062 0.4
rs4963128 (KIAA1542) 30.3 (955/3,150) 34.0 (1,173/3,448) 0.84 (0.8 to 0.9) 0.0011 0.2
rs6445975 (PXK) 27.3 (772/2,824) 24.2 (734/3,034) 1.19 (1.1 to 1.3) 0.0038 0.7
rs509749 (LY9) 43.1 (1,359/3,154) 43.8 (1,513/3,452) 0.97 (0.9 to 1.1) 0.5 0.4
Loci ordered by decreasing effect size (odds ratio (OR)). All results refer to the minor allele of each SNP, which is indicated in Table 1. CI,
confidence interval; SLE, systemic lupus erythematosus. aData presented as percentage (number of minor alleles/total number of alleles).
Page 4 of 9
(page number not for citation purposes)
94
Procedimiento experimental y resultados
Table 3
Combined analysis of allele frequency differences between SLE women and control women
Minor allele frequency (%)a Mantel – Haenszel analysis Breslow – Day test
rs1143679 (ITGAM) 22.3 (621/2,782) 15.1 (329/2,182) 1.67 (1.4 to 2.0) 2.0 × 10-11 0.6
rs7574865 (STAT4) 33.1 (636/1,920) 24.0 (317/1,322) 1.60 (1.4 to 1.9) 8.4 × 10-9 0.8
rs13277113 (C8orf13-BLK) 30.9 (777/2,514) 25.7 (475/1,848) 1.33 (1.2 to 1.5) 5.4 × 10-5 0.9
rs2304256 (TYK2) 22.9 (638/2,782) 27.2 (592/2,180) 0.81 (0.7 to 0.9) 0.0022 0.2
rs17435 (MECP2) 26.7 (744/2,784) 22.8 (498/2,186) 1.26 (1.1 to 1.4) 0.00085 0.7
rs10798269 (1q25.1) 27.1 (754/2,786) 32.3 (705/2,182) 0.77 (0.7 to 0.9) 6.2 × 10-5 0.4
rs17266594 (BANK1) 23.9 (464/1,938) 26.9 (386/1,436) 0.87 (0.7 to 1.0) 0.077 0.4
rs4963128 (KIAA1542) 30.4 (845/2,778) 33.9 (741/2,182) 0.85 (0.8 to 1.0) 0.011 0.2
rs6445975 (PXK) 26.7 (670/2,514) 23.5 (436/1,854) 1.22 (1.1 to 1.4) 0.0067 0.9
rs509749 (LY9) 43.3 (1,206/2,784) 44.0 (962/2,184) 0.98 (0.9 to 1.1) 0.7 0.5
Loci ordered as presented in Table 2. All results refer to the minor allele of each SNP, which is indicated in Table 1. CI, confidence interval; OR,
odds ratio; SLE, systemic lupus erythematosus. aData presented as percentage (number of minor alleles/total number of alleles).
describing association of the ITGAM gene it was indicated 0.03). Some SNPs were not associated in men (in the TYK2,
that results were not different between women and men [7]. 1q25.1, BANK1 and LY9 loci), but statistical power of this
This lack of information is probably due to the rarity of men suf- subgroup analysis was low, ranging from 0.19 for
fering from SLE. In our analysis, we have considered all male rs17266594 in BANK1 to 0.25 for rs2304256 in TYK2
data together without stratifying for sample collection due to among the nonassociated SNPs (power was estimated for P
the low number of men in each collection (Table 4). Results in = 0.05 and OR = 1.2).
men were similar to results in women, with the possible excep-
tion of the rs1143679 in ITGAM (OR = 2.08 versus 1.67; P =
Table 4
Comparison of SNP allele frequencies between SLE men and control men
rs1143679 (ITGAM) 27.9 (83/298) 15.7 (181/1,154) 2.08 (1.5 to 2.8) 1.2 × 10-6
rs7574865 (STAT4) 31.7 (64/202) 21.5 (159/740) 1.69 (1.2 to 2.4) 0.0025
rs13277113 (C8orf13-BLK) 31.0 (88/284) 25.1 (267/1,064) 1.34 (1.0 to 1.8) 0.045
rs2304256 (TYK2) 26.8 (80/298) 28.8 (334/1,158) 0.91 (0.7 to 1.2) 0.5
rs17435 (MECP2)b 29.2 (42/144) 18.5 (105/568) 1.82 (1.2 to 2.8) 0.0046
rs10798269 (1q25.1) 28.0 (84/300) 30.7 (355/1,158) 0.88 (0.7 to 1.2) 0.4
rs17266594 (BANK1) 26.2 (56/214) 28.4 (227/798) 0.89 (0.6 to 1.3) 0.5
rs4963128 (KIAA1542) 28.0 (84/300) 34.9 (403/1,156) 0.73 (0.6 to 1.0) 0.025
rs6445975 (PXK) 32.1 (86/268) 24.8 (265/1,068) 1.43 (1.1 to 1.9) 0.016
rs509749 (LY9) 40.6 (121/298) 42.9 (496/1,156) 0.91 (0.7 to 1.2) 0.5
Loci ordered as presented in Table 2. All results refer to the minor allele of each SNP, which is indicated in Table 1. No stratified analysis by
sample collection was done due to the small number of patients with systemic lupus erythematosus (SLE) in each collection. CI, confidence
interval; OR, odds ratio. aData presented as percentage (number of minor alleles/total number of alleles). bThese results are of carrier analysis
because MECP2 is in the X chromosome and there is a single allele in each man.
Page 5 of 9
(page number not for citation purposes)
95
Factores genéticos asociados con predisposición a LES
Following in importance is the association of MECP2 because We have also found a significant association with the
our results provide replication and indicate that a previous rs17266594 in the BANK1 gene. This SLE genetic factor has
assumption about the role of this genetic factor in contributing been identified in a low-resolution GWA study in a Swedish
to the sex bias in SLE is questionable. Sawalha and col- sample and replicated in other European sample collections in
leagues considered the X-chromosome methyl CpG binding the same study [5], but it was not found in any of the high-res-
protein 2 coding gene (MECP2) as a possible SLE genetic olution GWA studies and has not yet been replicated by other
factor based on two features: SLE predominance in women groups. Our results provide this independent replication,
and abnormal regulation of methylation-sensitive T-cell genes although with a more modest effect (OR = 0.83 in our study
Page 6 of 9
(page number not for citation purposes)
96
Procedimiento experimental y resultados
versus 0.70 in Kozyrev and colleagues [5]). The causal poly- In addition, we have found that most examined SLE-associ-
morphism can be the rs17266594 SNP itself, which seems to ated SNPs seem to be shared between women and men.
alter splicing efficiency of BANK1, or two BANK1 nonsynon- Results are not definitive given the small number of men in the
ymous SNPs of possible damaging effect. Linkage disequilib- patient group. This lack of differential association is important
rium between these three SNPs has prevented dissection of because we do not know definitively the causes of the female
their relationship to SLE susceptibility [5]. BANK1 codes for a preference of SLE. Lack of detailed gender analysis in previ-
B-cell scaffold protein with ankyrin repeats that is implicated in ous genetic reports is regrettable because only aggregation of
B-cell receptor-mediated signalling. data from multiple studies will allow us to know whether
genetic factors contribute to this sex bias.
The rs509749 SNP of LY9 is the only SNP that was not repli-
cated in our study. We selected this SNP because it seems to Conclusions
explain the 1q23 SLE-linked locus according to a large family- In summary, our study has provided independent replication of
based study [14]. 1q23 is one of the most consistently nine SLE-associated loci, six of them of confirmatory impor-
described SLE loci in linkage studies (and its syntenic region tance because they have not yet been independently repli-
in the mouse lupus models) [1]. Examination of SNPs all along cated by other groups (1q25.1, MECP2, KIAA1542, PXK and
this locus showed stronger association with the rs509749 BANK1) or because their association was controversial
SNP [14]. This SNP has a predictable impact in protein func- (TYK2). These results bring the number of strongly confirmed
tion and is associated with changes in the proportion of spe- associated loci to 13. Replication in independent studies is
cific T-cell subsets [14]. All this evidence made the rs509749 indispensable for considering a genetic factor in this category,
SNP a good candidate for replication in our view, even if the although the common use of multiple case–control sets inside
level of significance of the SLE association was notably lower the same study or of large sample collections has increased
than the reported for the other nine SNPs studied here (P = the chances of replication [2]. Some other promising associa-
0.002). Lack of replication of this SNP in contrast with replica- tions have been discovered [6,25], or await sufficient inde-
tion of the other nine SNPs provides support for the direct rela- pendent replication [2], but it is already certain that the genetic
tionship between very low P values obtained in sound studies component of SLE is especially rich in genetic factors with
and the reproducibility of genetic association findings [21]. effects above the detectable level with current studies (OR =
1.15 to 1.25). We are therefore now in a phase of exciting dis-
The most associated SNPs in our samples were the three that coveries in this field. There still remain formidable challenges,
were already confirmed previous to our study. These three however, because it is necessary to transform the information
SNPs were associated with SLE in at least three large studies. we obtain into useful knowledge and, as has been discussed
The largest effect was observed with a nonsynonymous SNP above, we have very few clues regarding the meaning of the
in the third exon of the ITGAM gene (rs1143679, OR = 1.70) identified SLE associations. Future studies should try to iden-
[3,4,6,7]. This nonsynonymous SNP was the most associated tify the causal variants and to determine their effect at molec-
in one of the previous studies (with very similar effect, OR = ular, cellular and disease levels, including the assessment of
1.74) [7], and has been hypothesized to disturb ITGAM inter- their role in the different SLE phenotypes and the probable
action with its ligands, but still no functional evidence is avail- similar effect in women and men.
able. Another clearly established association [3,4,6,8-12] was
the second strongest in our study: SNP rs7574865 in the Competing interests
third intron of the STAT4 gene (OR = 1.62). This association The authors declare that they have no competing interests.
seems stronger in patients with a severe phenotype [12]; how-
ever, no functional polymorphism has been identified in this Authors' contributions
locus. The next strongest association (OR = 1.34) was with MS-G participated in design of the study, in genotyping the
the rs13277113 SNP, which has been reported in the GWA samples, in interpretation of the results and in writing the man-
study of Hom and colleagues [4], with a similar effect (OR = uscript. MC participated in the statistical analysis and in the
1.39). This SNP is located between C8orf13 (of unknown interpretation of results. EE, RP, JO-R, GDS, SR, MJS, CP,
function) and BLK (B-lymphoid tyrosine kinase), two genes MM, FNS, AS, FJB, SD'A, MB, PC, TW and SM participated
that are transcribed in opposite directions. No functional vari- in the acquisition of clinical data and collection of samples and
ant has been identified in this locus, but the risk allele of this in the analysis and interpretation of results. JJG-R coordinated
SNP correlates with low mRNA levels of BLK and high levels the acquisition of clinical data and collection of samples and
of C8orf13, raising the possibility that either of these two participated in the analysis and interpretation of results. AG
effects could be related with SLE. Graham and colleagues participated in the design of the study and in the coordination
found association with a strongly linked SNP in the BLK gene of acquisition of clinical data and collection of samples, and
[6], while the SLEGEN GWA study found association with an supervised genotyping, statistical analysis, interpretation of
unlinked SNP in this locus, suggesting the possibility of two results and writing of the manuscript. All authors read and
independent genetic factors [3]. approved the final manuscript.
Page 7 of 9
(page number not for citation purposes)
97
Factores genéticos asociados con predisposición a LES
Page 8 of 9
(page number not for citation purposes)
98
Procedimiento experimental y resultados
Page 9 of 9
(page number not for citation purposes)
99
100
Supplementary Table 1
Origin and female percentage of the DNA sample collections used for replication of SLE associated SNPs.
m
Spain, Barcelonam 96 89 52.1 91 Hospital Val d’Hebron. Barcelona
n n
Spain, Coruña 81 88 71.6 92 CH Universitario Juan Canalejo. Coruña
o
Spain, Madrido 98 92 64.3 91.3 Hospital 12 de Octubre. Madrid
p
Spain, Santiagop 95 109 48.4 89 Hospital Clinico Universitario de Santiago
Total 1726 1579 63.2 88.2
Procedimiento experimental y resultados
Supplementary Table 2
Clinical characteristics of the patients with SLE included in the study
101
102
Supplementary Table 3
Primers and probes in 5’ to 3’ direction used for genotyping the 10 SNPs by the SNaPshot technique
PCR primers
Locus SNP Forward Primer Reverse Primer Minisequencing Probe
C8orf13-BLK rs13277113
TTACCTATGATTGATCGTGGTGATATCCGtctgacgcaggcc
cattaggtaacctgtgcttttcct ccagtccaagattcacctcag
atttttaagattaaacacttatcagatcatt $
KIAA1542
TGATTGATCGTGGTGATATCCGcctgcctccccgagtgtgtc
rs4963128 ccgagatcttgactccgtccg ggaggacaggaaggcaccaag
c $
1q25.1 rs10798269
AGAAGTATATTAATGAGCAGTGCAGttgaggtcgaccaatct
cactaaggatggaccatttgagg aggccatggctggggaga
ctcctccttttctccttacaaatcta $
BANK rs17266594
TCGTGGTGATATCCGtgcctagtgagcatattaaaatttttg
gcaaattaatatattgcctagtgagc acaggatggcttcccttttc
agaaataataatttaacctgc $
MECP2 rs17435
ATCCGtgtagtcttcagccctatctggattcttctctttgtt
gcccagtgagccagcccag cggagagaatttgcacgctg
ctaagaactgtgttgg $
Factores genéticos asociados con predisposición a LES
LY9 rs509749
ACCTTCGCGGCAGATATAATtccatatgtcacggaagttgag
tcactcaggagggcgctgg tgcccgtatgcccctagtc
tctgtggttggagagaacacc $
STAT4 rs7574865
catgagtgtgtatgcagtaaaagtatgaaaagttggtgagca
tggtgtggatggaggtaagg aatcccctgaaattccactga
aaatgt
Primers selected with Primer3 and Oligos softwares. They were checked to avoid formation of dimers between the primers included in the reaction.
$ These oligonucleotides were extended with a 5' tail that has no homology with human sequences (these tails are in capital letters)
Supplementary Table 4
Genotype counts for each of the ten SNPs and each of the sample collections
Data from sample collections that have been included in other publications were excluded (empty cells)
Data for the rs17266594 SNP from the Barcelona collection were excluded due to inconsistencies in regenotyping
Sample rs2304256 (TYK2) rs7574865 (STAT4) rs6445975 (PXK) rs17435 (MECP2) rs509749 (LY9) rs4963128 (KIAA1542) rs1143679 (ITGAM) rs13277113 (C8orf13-BLK rs17266594 (BANK1) rs10798269 (1q25.1)
collections CC CA AA GG GT TT TT TG GG AA AT TT AA AG GG GG GA AA GG GA AA GG GA AA TT TC CC GG GA AA
SLE 57 32 5 44 35 15 40 42 12 56 30 7 27 44 23 46 30 18 52 36 6 50 40 4 57 36 1 47 41 6
Portugal Controls 46 36 13 52 40 3 51 36 8 61 32 2 29 42 24 51 35 9 61 31 3 56 36 3 42 45 8 38 46 11
SLE 66 23 3 39 46 7 55 30 7 56 34 2 34 44 14 57 26 9 57 29 6 47 34 11 53 36 3 63 25 4
Madrid Controls 50 44 4 63 29 6 58 37 3 70 19 8 25 50 23 46 43 9 72 25 1 59 32 7 53 42 3 40 50 8
SLE 66 39 4 57 42 10 62 37 10 64 40 5 35 51 23 56 39 14 49 52 7 60 42 7 58 49 2 58 41 10
Santiago Controls 52 36 7 59 32 4 49 40 6 76 11 8 30 50 15 37 47 11 72 20 3 58 31 6 53 33 9 49 38 8
SLE 77 64 6 80 60 7 96 44 7 40 70 37 67 64 16 82 56 9 81 50 16 78 62 7
Asturias Controls 112 75 13 134 61 5 138 43 18 50 104 45 85 83 32 138 57 5 123 67 10 86 86 28
SLE 47 35 6 47 33 8 48 38 2 43 34 10 24 46 18 52 30 6 54 27 7 42 40 6 55 29 4 52 34 2
Coruña Controls 42 32 7 51 27 4 46 31 5 54 22 5 28 34 19 46 31 5 60 18 1 53 23 3 50 29 3 40 38 3
SLE 47 36 6 42 37 10 53 29 6 49 32 8 26 48 15 44 35 10 44 35 9 43 39 7 47 38 4
Barcelona Controls 58 27 11 52 38 6 57 30 9 63 25 8 29 48 19 53 33 10 66 25 5 55 38 3 47 41 8
SLE 46 35 3 44 26 14 28 45 11 32 38 14 49 29 6 45 33 6
Rome Controls 59 35 8 63 20 19 34 51 17 43 41 18 70 27 5 61 28 13
SLE 66 54 7 59 54 15 67 55 6 71 44 11 38 70 20 61 59 7 76 43 8 69 45 13 72 50 6 64 57 7
Milan Controls 56 40 10 64 36 6 71 30 5 76 18 12 38 51 17 43 41 22 70 32 4 60 40 6 58 39 9 53 41 12
SLE 47 27 7 40 30 11 31 41 9 38 30 13 40 35 6 47 28 6
Naples Controls 45 55 7 55 45 8 36 48 24 43 52 13 77 30 1 49 49 10
The SLE 61 36 5 47 41 16 64 26 13 37 45 20 55 39 10 63 38 3 30 58 15 58 40 6 49 45 10
Netherland Controls 94 71 15 92 73 15 127 29 21 65 88 27 90 78 12 130 48 2 63 95 22 98 68 14 74 82 24
SLE 57 35 3 48 39 8 46 39 10 47 38 10 32 51 12 52 33 10 64 30 1 45 38 12 46 38 11 48 44 3
Hungary Controls 50 41 4 58 35 2 46 46 3 67 17 10 28 38 29 37 45 13 77 17 1 55 35 5 48 39 8 42 42 11
SLE 69 25 3 38 48 11 58 36 3 49 31 17 28 54 15 44 40 13 52 40 5 47 39 11 53 38 6 47 39 11
Greece 1 Controls 47 43 9 50 43 6 60 36 2 63 25 9 37 42 20 31 54 14 62 34 2 54 38 6 43 47 9 50 39 10
SLE 59 27 8 36 43 15 50 39 5 40 40 14 19 56 19 51 33 10 47 43 4 40 39 15 63 27 4 51 37 6
Greece 2 Controls 54 27 6 54 29 5 59 25 4 44 37 5 30 34 24 29 45 12 58 28 2 37 41 9 42 40 6 36 43 8
SLE 58 28 8 41 41 12 39 47 8 57 26 11 35 45 14 46 39 9 73 20 1 43 46 5 52 32 10 55 31 8
Slovakia Controls 47 38 8 58 30 5 42 41 10 55 26 12 32 51 10 41 42 10 75 16 2 57 33 3 46 39 8 51 32 10
Czech SLE 62 32 7 43 47 11 54 40 7 55 34 12 38 37 26 53 39 9 75 25 1 45 48 8 60 37 4 49 37 15
Republic Controls 46 45 8 52 42 5 59 31 9 72 11 16 36 50 13 43 40 15 77 22 0 54 40 4 58 33 8 47 44 8
SLE 45 31 6 44 33 4 47 30 3 29 46 7 41 31 7 53 24 5 38 32 12 32 41 9
Germany Controls 45 39 8 51 33 8 66 22 4 32 40 20 49 31 11 75 17 0 49 33 10 45 39 8
SLE 930 559 87 494 465 122 743 566 103 878 539 155 501 793 283 795 605 175 930 562 84 680 590 142 627 412 57 832 633 114
Total Controls 903 684 138 613 381 52 875 550 92 1150 402 165 559 821 346 767 741 216 1240 447 37 833 582 97 591 454 85 808 738 180
103
Procedimiento experimental y resultados
Supplementary Table 5
104
Minor allele frequencies for each of the ten SNPs and each of the sample collections
Data from sample collections that have been included in other publications were excluded (empty cells)
Data for the rs17266594 SNP from the Barcelona collection were excluded due to inconsistencies in regenotyping
rs2304256 rs7574865 rs6445975 rs17435 rs509749 rs4963128 rs1143679 rs13277113 rs17266594 rs10798269
(TYK2) (STAT4) (PXK) (MECP2) (LY9) (KIAA1542) (ITGAM) (C8orf13-BLK) (BANK1) (1q25.1)
Sample collections A T G T G A A A C A
SLE 0.223 0.346 0.351 0.237 0.479 0.351 0.255 0.255 0.202 0.282
Portugal Controls 0.326 0.242 0.274 0.189 0.474 0.279 0.195 0.221 0.321 0.358
SLE 0.158 0.326 0.239 0.207 0.391 0.239 0.223 0.304 0.228 0.179
Madrid Controls 0.265 0.209 0.219 0.180 0.490 0.311 0.138 0.235 0.245 0.337
SLE 0.216 0.284 0.261 0.229 0.445 0.307 0.306 0.257 0.243 0.280
Santiago Controls 0.263 0.211 0.274 0.142 0.421 0.363 0.137 0.226 0.268 0.284
SLE 0.259 0.252 0.197 0.490 0.327 0.252 0.279 0.259
Asturias Controls 0.253 0.178 0.198 0.487 0.368 0.168 0.218 0.355
SLE 0.267 0.278 0.239 0.310 0.466 0.239 0.233 0.295 0.210 0.216
Coruña Controls 0.284 0.213 0.250 0.198 0.444 0.250 0.127 0.184 0.213 0.272
SLE 0.270 0.320 0.233 0.270 0.438 0.309 0.301 0.298 0.258
Barcelona Controls 0.255 0.260 0.250 0.214 0.448 0.276 0.182 0.229 0.297
SLE 0.244 0.321 0.399 0.393 0.244 0.268
Rome Controls 0.250 0.284 0.417 0.377 0.181 0.265
SLE 0.268 0.328 0.262 0.262 0.430 0.287 0.232 0.280 0.242 0.277
Milan Controls 0.283 0.226 0.189 0.198 0.401 0.401 0.189 0.245 0.269 0.307
SLE 0.253 0.321 0.364 0.346 0.290 0.247
Naples Controls 0.322 0.282 0.444 0.361 0.148 0.319
SLE 0.225 0.351 0.252 0.417 0.284 0.212 0.427 0.250 0.313
The Netherlands Controls 0.281 0.286 0.201 0.394 0.283 0.144 0.386 0.267 0.361
Factores genéticos asociados con predisposición a LES
SLE 0.216 0.289 0.311 0.305 0.395 0.279 0.168 0.326 0.316 0.263
Hungary Controls 0.258 0.205 0.274 0.197 0.505 0.374 0.100 0.237 0.289 0.337
SLE 0.160 0.361 0.216 0.335 0.433 0.340 0.258 0.314 0.258 0.314
Greece 1 Controls 0.308 0.278 0.204 0.222 0.414 0.414 0.194 0.255 0.328 0.298
SLE 0.229 0.388 0.261 0.362 0.500 0.282 0.271 0.367 0.186 0.261
Greece 2 Controls 0.224 0.222 0.188 0.273 0.466 0.401 0.182 0.339 0.295 0.339
SLE 0.234 0.346 0.335 0.255 0.388 0.303 0.117 0.298 0.277 0.250
Slovakia Controls 0.290 0.215 0.328 0.269 0.382 0.333 0.108 0.210 0.296 0.280
SLE 0.228 0.342 0.267 0.287 0.441 0.282 0.134 0.317 0.223 0.332
Czech Republic Controls 0.308 0.263 0.247 0.217 0.384 0.357 0.111 0.245 0.247 0.303
SLE 0.262 0.253 0.225 0.366 0.285 0.207 0.341 0.360
Germany Controls 0.299 0.266 0.163 0.435 0.291 0.092 0.288 0.299
Procedimiento experimental y resultados
Lack of Interaction Between Systemic Lupus by its proxy, rs2070197) and rs729302. The first SNP showed the strongest
Erythematosus-associated Polymorphisms in TYK2 and IRF5 association (p = 4.8 × 10–20) with SLE in our study8 and others10 and
accounts for association of many other SNP in the gene, including
To the Editor: rs109542138,10. The second, rs729302, is an SNP in the promoter region of
These are exciting times for the genetic investigation of systemic lupus IRF5 that was independently associated to SLE in our samples (p = 1.6 ×
erythematosus (SLE), characterized by the discovery of many reproducibly 10–9). Neither of the 2 SNP showed evidence of interaction with the
associated loci1. Further progress will require research in many different rs2304256 SNP of TYK2, either with an analysis as done by Hellquist, et
directions, including investigation of how the effects of each locus inte- al2 or with LRASSOC (Table 1). These comparisons are more powerful
grate between them and with environmental exposures to cause SLE. We than models with rs10954213 because the 2 IRF5 SNP are much more
read with interest the report by Hellquist, et al2 that showed evidence of strongly associated to SLE susceptibility and because we have data for
significant epistatic interaction between 2 SLE-associated loci, IRF5 and 3-fold more samples: 1223 cases and 1300 controls.
TYK2. The first is a definitively confirmed SLE susceptibility locus with Our results did not show any statistical evidence of interaction between
one of the strongest, albeit complex, effects. The latter has been more con- SLE-associated SNP in TYK2 and IRF5. As a consequence, we should con-
tentious, but its association with SLE is becoming clearer2,3. Epistasis tinue to consider that these 2 SLE genetic factors can contribute to disease
means that risk in subjects with susceptibility alleles at the 2 loci signifi- susceptibility by independent pathways, unless other genetic studies repli-
cantly exceeds the sum of the risks at each locus. This was rightly inter- cate the epistatic interaction or functional studies support the interaction.
preted to mean that the 2 loci impinge in the type 1 interferon pathway2, Similar interaction analysis for other genetic factors and in other sample
which is an important insight because TYK2 codes for a Janus kinase that collections will unravel the influence of epistatic interaction in SLE sus-
is involved in multiple cytokine signaling pathways in addition to this one4. ceptibility, but current evidence does not support an important contribu-
Also, demonstration of epistasis between 2 SLE loci is of importance tion5 (Suarez-Gestal, et al, unpublished data).
because its absence has been the rule for SLE genetic factors5 and for most
genetic factors of other complex diseases6,7. What is common is the con- MARIAN SUAREZ-GESTAL, MSc; MANUEL CALAZA, MMath;
tribution to total risk of the different disease alleles in a purely additive ANTONIO GONZALEZ, MD, PhD; European Consortium of SLE DNA
way, without any interaction. Therefore, we were interested in replicating Collections, Laboratorio de Investigacion 10 and Rheumatology Unit,
the epistatic interaction described by Hellquist, et al2 with the SLE data Hospital Clinico Universitario de Santiago, Santiago de Compostela,
available to us. Spain. Address correspondence to Dr. Gonzalez;
We had already genotyped the relevant single-nucleotide polymor- E-mail: [email protected]
phisms (SNP), rs2304256 in TYK23 and rs10954213 in IRF58, in a large
collection of patients with SLE and controls. These 2 SNP were associated REFERENCES
with SLE in our studies (p = 2.5 × 10–5 and 0.015, respectively) with risk 1. Rhodes B, Vyse TJ. The genetics of SLE: an update in the light of
alleles that were coincident to those found by Hellquist, et al (who report- genome-wide association studies. Rheumatology 2008;47:1603-11.
ed p = 10–4 and p = 0.004, respectively). We have data for the 2 SNP in 419 2. Hellquist A, Järvinen TM, Koskenmies S, Zucchelli M,
SLE subjects and 454 controls. The same type of analysis done by Orsmark-Pietras C, Berglind L, et al. Evidence for genetic
Hellquist, et al2, that is, a comparison between the fit to the data of logis- association and interaction between the TYK2 and IRF5 genes in
tic regression models with and without an interaction term, showed no dif- systemic lupus erythematosus. J Rheumatol 2009;36:1631-8.
ferences (Table 1). As a more exhaustive and sensitive test, we also con- 3. Suarez-Gestal M, Calaza M, Endreffy E, Pullmann R, Ordi-Ros
ducted analysis with the LRASSOC software9. This approach compares a J, Domenico Sebastiani G, et al. Replication of recently identified
range of specific genetic models with and without interaction and does not systemic lupus erythematosus genetic associations: a case-control
require statistical significance to discriminate between models, but only study. Arthritis Res Ther 2009;11:R69.
differences according to the less stringent Akaike’s Information Criterion 4. Minegishi Y, Saito M, Morio T, Watanabe K, Agematsu K, Tsuchiya
(AIC). The best model has the lowest AIC value, which means that it has S, et al. Human tyrosine kinase 2 deficiency reveals its requisite
the best fit to the case-control genotypes with the highest combination of roles in multiple cytokine signals involved in innate and acquired
likelihood and parsimony. These analyses indicated that the best model immunity. Immunity 2006;25:745-55.
included the independent contribution of the 2 loci without any interaction, 5. International Consortium for Systemic Lupus Erythematosus
although models with interaction terms were not much worse (Table 1). Genetics (SLEGEN), Harley JB, Alarcón-Riquelme ME, Criswell
Therefore, we did not confirm the epistatic interaction that Hellquist and LA, Jacob CO, Kimberly RP, Moser KL, et al. Genome-wide
colleagues have described. association scan in women with systemic lupus erythematosus
As a complementary analysis, we checked whether there was statistical identifies susceptibility variants in ITGAM, PXK, KIAA1542 and
evidence of interaction between TYK2 and IRF5 using other SNP in IRF5. other loci. Nat Genet 2008;40:204-10.
These SNP were rs10488631 (which in some studies10 has been replaced 6. Wellcome Trust Case Control Consortium. Genome-wide
Table 1. Lack of epistatic interaction between the SLE-associated SNP in TYK2 and IRF5.
* Multivariate logistic regression models with and without an interaction term were compared as in Hellquist, et al2. The models did not specify any mode
of inheritance. P values were obtained with the likelihood ratio test. ** Comparison of the specific inheritance models included in LRASSOC9. Akaike’s
Information Criterion (AIC) was used to classify the models. A value of AIC > 2 indicates a meaningful difference. † Additive: model with additive effects
of each SNP, without dominance or interactive components. SNP: single-nucleotide polymorphisms.
105
Factores genéticos asociados con predisposición a LES
association study of 14,000 cases of seven common diseases and 9. North BV, Curtis D, Sham PC. Application of logistic regression to
3,000 shared controls. Nature 2007;447:661-78. case-control association studies involving two causative loci. Hum
7. Nair RP, Duffin KC, Helms C, Ding J, Stuart PE, Goldgar D, et al. Hered 2005;59:79-87.
Genome-wide scan reveals association of psoriasis with IL-23 and 10. Kozyrev SV, Lewén S, Reddy PM, Pons-Estel B, Argentine
NF-kappa-B pathways. Nat Genet 2009;41:199-204. Collaborative Group, Witte T, et al. Structural insertion/deletion
8. Ferreiro-Neira I, Calaza M, Alonso-Perez E, Marchini M, Scorza R, variation in IRF5 is associated with a risk haplotype and defines the
Sebastiani GD, et al. Opposed independent effects and epistasis in precise IRF5 isoforms expressed in systemic lupus erythematosus.
the complex association of IRF5 to SLE. Genes Immun Arthritis Rheum 2007;56:1234-41.
2007;8:429-38.
J Rheumatol 2010;37:3; doi:10.3899/jrheum.090823
Correspondence 677
106
2. Estudio funcional de PD1.3
Procedimiento experimental y resultados
ORIGINAL ARTICLE
Analysis of the functional relevance of a putative
regulatory SNP of PDCD1, PD1.3, associated
with systemic lupus erythematosus
M Suarez-Gestal1,4, I Ferreiros-Vidal1,4, JA Ortiz2, JJ Gomez-Reino1,3 and A Gonzalez1
1
Laboratorio de Investigacion 2 and Rheumatology Unit, Hospital Clinico Universitario de Santiago, Santiago de Compostela, Spain;
2
Instituto Bernabeu, Departamento de Biologı́a Molecular, Alicante, Spain and 3Department of Medicine, University of Santiago de
Compostela, Santiago de Compostela, Spain
This study aimed to test the functional effects of the PD1.3 single nucleotide polymorphism (SNP) (rs11568821), which were
proposed based on its association to systemic lupus erythematosus (SLE) susceptibility and in electrophoretic mobility shift
assays (EMSA) results. We analysed transcriptional effects of the PD1.3 locus by enhancer reporter assays. Results were
against the hypothesis that the PD1.3 locus acts as enhancer in transcriptional regulation of PDCD1. In addition, they excluded
a differential effect of the PD1.3 alleles. EMSA results confirmed that oligonucleotides with the PD1.3 G allele bind RUNX1 but
not those with the A allele. However, binding to PD1.3 G oligonucleotides was much lower than binding to positive control
oligonucleotides. Criss-cross experiments showed that this was due to flanking nucleotides in the PD1.3 sequence that
negatively affect RUNX1 binding. These results cast doubts on the functional relevance of the PD1.3 SNP and, together with
the lack of association in several studies, put into question its role as an SLE susceptibility factor. Investigation of other PDCD1
polymorphisms is needed to uncover the possible effect of this gene on SLE susceptibility.
Genes and Immunity advance online publication, 10 April 2008; doi:10.1038/gene.2008.19
Keywords: systemic lupus erythematosus; functional polymorphism; transcriptional regulation; transcription factor
binding site
109
Factores genéticos asociados con predisposición a LES
110
Procedimiento experimental y resultados
Figure 1 Alignment of the tandem repeat structure near the PD1.3 SNP (encased) in PDCD1 intron 4 with ortologue mammalian genome
sequences. The plus strand is presented although the PDCD1 gene is transcribed in the minus strand (G and A alleles in place of the C and T
presented here). Spaces are adjusted to highlight tandem repeat structure: two 80 bp repeats and partly overlapping four 40 bp repeats. The
latter are indicated by alternating lower and upper case and separated by spaces. Some potential transcription factor binding sites in the
minus strand of PDCD1 have been signalled.
111
Factores genéticos asociados con predisposición a LES
C fos
fos luc
luc
*
fos
G fos luc
luc
*
fos
fos luc
luc C
fos
fos luc
luc G
fos
fos luc
luc
0 2 4 6 8 10 12 14 0 2 4 6 8 10 12 14 16
RLU RLU
Figure 2 Luciferase enhancer reporter assays exploring the tandem repeat region of PDCD1 intron 4 containing the PD1.3 SNP. Vectors
containing this region in the forward or reverse orientation and upstream or downstream of a Firefly Luciferase gene (luc) under control of
a minimal fos promoter, were compared with the activity of a vector without it in Jurkat cells (a) in basal conditions or (b) after phorbol
12-myristate 13-acetate and ionomycin stimulation. These graphs represent the mean and s.e. of five independent experiments. *Po0.05.
@ fos
fos luc
@ fos luc
fos
fos luc @
fos
fos luc @
@
0.6 0.8 1.0 1.2 1.4 1.6 1.8 2.0 2.2 2.4 0.4 0.7 1.0 1.3 1.6 1.9 2.2 2.5 2.8
Figure 3 Lack of difference between the G and A allele at the PD1.3 SNP of PDCD1 in luciferase enhancer reporter assays. The two PD1.3
alleles were compared in transiently transfected Jurkat cells: (a) in basal conditions and (b) after phorbol 12-myristate 13-acetate and
ionomycin stimulation. Vectors were as in Figure 2 but either with the G or with the A allele at PD1.3 (their position represented by @).
Results are geometric means and 95% confidence interval of the ratio between luciferase signal in cells transfected with the A allele and cells
transfected with the G allele in homologous vectors. Five independent experiments were analysed.
112
Procedimiento experimental y resultados
hexanucleotide showed much stronger binding at the same nuclear nucleotides, two on each side, flanking the central hexanucleotide
extract concentration (lane 1 versus 3). The need to use large from the PD1.3 site. It also compares binding to a PD1.3
amounts of nuclear extracts and long exposure times to visualize oligonucleotide, P12, and its modified version, Pm 12 containing the
P18G binding led to increased background and unspecific bands, and four flanking nucleotides from C1. Binding was much lower to Cm 1
to saturation of the stronger bands that prevented quantitative (lane 4) than to C1 (lane 1), and much stronger to Pm
12 (lane 8) than P12
113
Factores genéticos asociados con predisposición a LES
114
Procedimiento experimental y resultados
115
3. Influencia del componente genético en la
variabilidad clínica del LES
Procedimiento experimental y resultados
Marian Suarez-Gestal1, Manuel Calaza1, Myriam Liz1, Josep Ordi-Ros2, Eva Balada2,
Marc Bijl3, Cees G Kallenberg3, Chryssa Papasteriades4, Iris Kappou-Rigatou4, Patricia
Carreira5, Fotini N. Skopouli6, Maria Mavromati6, Reinhold E. Schmidt7, Torsten
Witte7, Emöke Endreffy8, Attila Kovacs9, Maurizio Marchini10, Raffaella Scorza10,
Sergio Migliaresi11, Gian Domenico Sebastiani12, Maria Jose Santos13, Filipe Vinagre13,
Ana Suarez14, Carmen Gutierrez14, Ignacio Rego15, Francisco J Blanco15, Nadia
Barizzone16, Sandra D’Alfonso16, Rudolf Pullmann Jr17, Rudolf Pullmann18, Sarka
Ruzickova19, Ctibor Dostal20, Juan J Gomez-Reino1,21 and Antonio Gonzalez1
1
Laboratorio de Investigacion 10 and Rheumatology Unit, Instituto de Investigacion
Sanitaria - Hospital Clinico Universitario de Santiago, Santiago de Compostela, 15706
Spain. [email protected]; [email protected]; [email protected]
2
Internal Medicine, Research Laboratory in Autoimmune Diseases, Hospital Vall
d'Hebron, 08035 Barcelona, Spain. [email protected]; [email protected]
3
Department of Rheumatology and Clinical Immunology, University Medical Center
Groningen, 9713 Groningen, The Netherlands. [email protected];
[email protected]
4
Department of Histocompatibility and Immunology, Evangelismos Hospital, 10676
Athens, Greece. [email protected]; [email protected]
5
Rheumatology Department, Hospital 12 de Octubre, 28041 Madrid, Spain.
[email protected]
6
Pathophysiology Department, Athens University Medical School, Athens 115 27,
Greece. [email protected]; [email protected]
7
Division of Clinical Immunology, Department of Internal Medicine of the Hannover
Medical School, D-30625 Hannover, Germany. [email protected];
[email protected]
8
Paediatrics Department, Albert Szent-Györgyi Medical and Pharmaceutical Centre,
University of Szeged, 6721 Hungary. [email protected]
9
Department of Rheumatology, Hospital of Hungarian Railways, H-5000 Szolnok,
Hungary. [email protected]
119
Factores genéticos asociados con predisposición a LES
10
Clinical Immunology, University of Milan and Fondazione IRCCS Ospedale
Maggiore Policlinico, Mangiagalli e Regina Elena, 20122 Milan, Italy.
[email protected]; [email protected]
11
Rheumatology Unit, Second University of Naples, 81100 Naples, Italy.
[email protected]
12
UOC Reumatologia, Azienda Ospedaliera San Camillo-Forlanini, Roma, Italy.
[email protected]
13
Rheumatology Department, Hospital Garcia de Orta and Rheumatology Research
Unit, Instituto Medicina Molecular, Lisboa, Portugal. [email protected];
[email protected]
14
Department of Functional Biology, Hospital Universitario Central de Asturias,
Universidad de Oviedo, Oviedo 33006, Spain. [email protected]; [email protected]
15
INIBIC-CH Universitario A Coruña, 15006 A Coruña, Spain. [email protected];
[email protected]
16
Dept Medical Sciences and IRCAD , Eastern Piedmont University, 28100 Novara,
Italy. [email protected]; [email protected]
17
GBMC Hospital, Department of Medicine, Baltimore, MD 21209, USA.
[email protected]
18
Institute of Clinical Biochemistry, Martin Faculty Hospital, Jessenius Medical
Faculty, Kollárova 2, 036 59 Martin, Slovakia. [email protected]
19
Institute of Biotechnology, Academy of Sciences of the Czech Republic, Prague,
Czech Republic. [email protected]
20
Molecular Biology and Immunogenetics Dept, Institute of Rheumatology, 128 50
Prague 2, Czech Republic. [email protected]
21
Department of Medicine, University of Santiago de Compostela, Santiago de
Compostela, 15706 Spain. [email protected]
Corresponding author:
Antonio Gonzalez
Laboratorio de Investigacion 10
Hospital Clinico Universitario de Santiago
Travesia de Choupana sn.
15706-Santiago de Compostela
Spain
120
Procedimiento experimental y resultados
121
Factores genéticos asociados con predisposición a LES
ABSTRACT
Introduction: To analyze if nine newly identified SLE susceptibility loci were
influencing the presence of some of the best defined SLE clinical features.
KIAA1542, PXK and 1q25.1, between patients with and without each of 11 clinical
features: ten of the ACR classification criteria (except ANAs) and age of disease onset.
Results: The most consistent and significant result was the association of the rare allele
of rs13277113 in the C8orf13-BLK locus with increased risk of nephritis (O.R. = 1.35, P
= 0.00026). The reported association of STAT4 with early age of disease onset was
replicated. Not so the other described associations of this locus, but joint analysis of a
previous report with our data lent further support to association of STAT4 with oral
ulcers and immunologic disorder. Combined analysis of ITGAM data from a recent
report and our study showed more doubtful association of this locus with immunologic
disorder, nephritis and discoid rash, although our study, separately, did not replicate
these associations. Some other results were suggestive of the presence of additional
phenotype-genotype associations, but they were not significant after correction for the
number of tests.
Conclusions: Our results together with previous studies indicate that clinical variability
of SLE is influenced by the SLE susceptibility alleles. However, the effects are weak
122
Procedimiento experimental y resultados
INTRODUCTION
understood [1]. Clinical heterogeneity complicates its diagnosis, treatment and research
[2]. We do not have yet clear clues about the causes of this variability, but likely
factors and the SLE features will help elucidate disease mechanisms and could provide
SLE clinical features have been reported, but they have not yet reached a high degree of
confidence [3-6]. This is similar to what had happened in relation to SLE susceptibility
until 2008: although there were many reports of association there were few genetic
factors that had been consistently demonstrated. The advent of Genome Wide
Association (GWA) studies in 2008 and other large studies, with their increased sample
sizes and stricter standards, changed dramatically this panorama. Now, there are more
than ten new loci that are consistently associated with SLE susceptibility [7-8]. These
loci are prime candidates for having a predisposing effect for specific SLE clinical
features. The first analyses seem to indicate that this is the case for some of them [9-11].
Herein, we have analyzed the association of nine of these consistent SLE susceptibility
and 1q25.1) with some clinical features in more than 1500 patients. Some significant
associations were found and some of the previously described received some additional
support, but the observed effects were weak and will need further confirmation.
123
Factores genéticos asociados con predisposición a LES
Sample and data collection: Recruiting of DNA samples from SLE patients has been
already described [12]. Briefly, 16 recruiting centres from 9 European countries were
asked for about 100 SLE patients and controls of uniform Caucasian ethnicity. A total
of 1561 patients were obtained (18 patients included in a previous study[12] were
excluded after reviewing their clinical data). All SLE patients met the revised American
consent. Sample collection was approved by the respective ethical committees. Clinical
characteristics of the patients were obtained at the same time. They included the ACR
IRAK1-MECP2, BANK1, TYK2, KIAA1542, PXK and 1q25.1 loci have been already
described [12].
Association analysis: We compared SNP allele frequencies between cases positive and
negative for each of the ACR classification criteria (except presence of ANA given that
they were almost constant) and age of disease onset by logistic regression following a
genetic additive model with codes 2 for aa genotypes, 1 for aA genotypes and 0 for the
AA genotypes. Patient gender was included as covariate. Age of disease onset was
included in these analysis after separating patients in two groups < or ≥ 30 years old at
disease onset. Results at this stage with P < 0.05 were analyzed in patients stratified by
gender. Bonferroni correction for multiple tests was done considering the number of
analyzed clinical features. Statistical analyses were done in a customized version of the
Statistica 7.0 program (StatSoft, Tulsa, OK, USA). Power to detect association was
estimated with the “Power and sample size” software [14]. Combination of the current
124
Procedimiento experimental y resultados
results with previous studies was done with fixed-effect and with random-effect
RESULTS
loci were available for 1561 European patients with SLE and for 1728 healthy controls
(Table 1). Information about ACR classification criteria, gender, age of disease onset,
and time of follow-up was also available for most of these patients (Table 2).
Seven associations between genotypes and clinical features were detected below
P = 0.05 in the comparison between positive and negative patients (Table 3). Only two
of them were below P = 0.01. The two involved nephritis that was associated with the
SLE risk allele at the C8orf13-BLK locus and with the IRAK1-MECP2 risk allele. The
other five associations were just below the 0.05 threshold. One of the weak associations
replicates a previous finding: the association of the risk allele of the STAT4 SNP with
younger disease onset [9]. However, we did not replicate the described associations of
the STAT4 SNP with nephritis, immunologic disorder or oral ulcers (although there was
a trend in the same direction for the latter) [9-10]. Other weak association was observed
between the ITGAM risk allele and a younger age of disease onset, but we did not
replicate other reported associations with this SLE risk allele [11]. We also found weak
association of renal disorder with the risk allele in BANK1 and of oral ulcers with this
same allele and the risk PXK allele (Table 3). However, only the association of nephritis
with the C8orf13-BLK SNP remained significant after correcting by the number of tests.
125
Factores genéticos asociados con predisposición a LES
Analyses of patients stratified by gender was done only for the SNPs showing P
< 0.05 in global analyses (Table 4). The smallest stratum included the 151 men with
SLE in our collection, what allowed for a very limited statistical power. This limitation
was considered for interpretation by focusing in effect sizes more than in P values. In
contrast, the largest stratum was made of 1393 women with SLE, and therefore similar
to the global study sample. The effect size of five of the associations was similar or
included the two stronger, renal disorder with the risk alleles of C8orf13-BLK and of
IRAK1-MECP2. The other two significant associations in the global analysis, renal
disorder with BANK1 and oral ulcers with PXK, were restricted to women in the
We combined our results with the available in previous reports. There were few
articles and only in three of them data were reported as to permit case-only analyses,
comparing patients with and without the clinical feature. Each of these three articles
provided data on one of the loci included in our analyses, C8orf13-BLK, STAT4 and
ITGAM [9, 11, 15]. There was an additional study analyzing SLE clinical features in
relation with the ITGAM locus but with a different SNP [15]. The other published
studies did not provide enough information to combine effect sizes, as a second one
about STAT4 [10], or the studies on BANK1 [16], and TYK2 [17]; or there was not any
renal disorder, was opposed to the results of a GWAS that included clinical data on 781
patients [15]. The marked contrast between the two studies leaves this association as
uncertain at this time (Table 5). On the contrary, the results for the STAT4 locus were
126
Procedimiento experimental y resultados
quite similar between the two available studies. The associations between young age of
disease onset, oral ulcers or immunologic disorder and rs7574865 were concordant and,
consequently, they became reinforced in the combined analysis with about 2700
patients (Table 5). Only the association that was previously observed between
rs7574865 and renal disorder did not receive any additional support, and it became non-
significant (P = 0.14) in the combined analysis. Finally, less conclusive results were
obtained with the joint analysis on the ITGAM locus. The direction of change for the
three analyzed criteria was the same in the two studies, but results from Kim-Howard et
al. showed stronger differences than ours. The differences in effects for renal disorder
and discoid rash were such as to cause conflicting results between the fixed-effects and
analysis and lack of them with the more conservative random-effects approach.
DISCUSSION
There are several obstacles for the identification of loci preferentially associated
with particular SLE phenotypes that make their investigation more difficult than the
study of disease susceptibility. Firstly, the statistical power of the available collections
becomes greatly reduced in subgroup analyses. Lack of power will lead to false negative
results. In addition, the performance of many comparisons increases the risk of false
positives and the subsequent lack of reproducibility. A solution to this problem will be
to require stringent significant thresholds for claiming association, but this measure will
lead to even more false negative results. Another limitation is the variability of the
clinical data, because sample collections show marked differences in the prevalence of
the different SLE manifestations [9, 15, 18-23]. These differences have many possible
127
Factores genéticos asociados con predisposición a LES
causes, but if they are due to biases unrelated with the disease process they will be a
recommendations for subgroup analysis in clinical trials [25-27], have determined the
design of our study. We have followed a fixed sequence approach, which requires a
significant difference in the more general group before starting analysis in a subgroup;
we have also corrected for the number of tests done, took into consideration previous
reports concerning the same comparisons, and take into account the power of the
According to the fixed sequence approach, we selected nine loci that are
convincingly associated to SLE susceptibility and that have shown association in our
collection of samples [7-8, 12]. This procedure does not exclude that some gene variants
susceptibility to SLE overall. This has been already reported for the FCGR3A V158F
SNP that is associated with lupus nephritis, but not convincingly with SLE
The clearest associations in our study were with renal disorder. Association
between lupus nephritis and the SLE risk allele of the rs13277113 SNP in the C8orf13-
BLK locus remained significant after correcting for the number of tests, and was
consistently observed in women and men. The same SNP has already been analyzed for
association with SLE classification criteria in a previous GWAS [15], but no association
with renal disorder was found in any of the two series of patients analyzed in that study.
In addition, the direction of change in one of the series, which was reported in more
detail, was opposed to the observed by us (Table 5). Prevalence of renal disorder in that
study was lower (28.0 %) than in our collection of samples (41.0 %) and this difference
(P = 2.0 x 10-12) could contribute to the discordant results. The C8orf13-BLK locus is
10
128
Procedimiento experimental y resultados
reproducibly associated with SLE susceptibility [12, 15, 28], and probably with RA [29-
30], and Systemic Sclerosis [31]. The SNP that we have analyzed is the most studied
and seems to be the most associated with SLE susceptibility. It correlates with changes
in expression of the two flanking genes, C8orf13 and BLK, with an increase of C8orf13
and a decrease of BLK [15]. BLK is the best understood of the two, and it is a good
candidate because it codes for a B-cell specific tyrosine kinase that is involved in
nephritis in our study was less clear. The SLE risk allele of rs17435 in the IRAK1-
MECP2 locus was associated with increased risk of nephritis both in women and men.
However, the association did not remain significant after correction by the number of
tests done. Therefore, we consider these two associations of renal disorder as unproven
The rs7574865 SNP in STAT4 has been associated with a severe SLE phenotype
defined by nephritis, age at diagnosis < 30 years old, immunologic disorder (and,
specifically, double-stranded DNA autoantibodies) and absence of oral ulcers [9]. The
nephritis prevalence was also reported in other study with 695 patients [10]. However,
no effect on age of disease onset or oral ulcers was detected in this second study. Our
results showed only a weak association with younger disease onset that was consistent
in women and men. We also found a trend to decreased prevalence of oral ulcers, but it
was only observed in women (data not shown). No association with nephritis or
immunologic disorder was present in our data. Overall, the results confirm that
rs7574865 SNP in the STAT4 locus is associated with a particular SLE phenotype
whose details are still incompletely defined, but that very likely include immunologic
disorder (same trend in the three studies), specially the presence of anti-dsDNA
11
129
Factores genéticos asociados con predisposición a LES
antibodies, which was a clear association in the two previous studies and was not
available to us; a younger age of disease onset, for which there was weak association in
two studies and a non-significant trend in the third [10]. It is also probable that this
phenotype will include also renal disorder and lack of oral ulcers because two of the
synonymous SNP in ITGAM, with a younger age of disease onset. This characteristic of
SLE has not been analyzed in previous studies [11, 15], and the association did not
remain significant after correction by the number of tests, which make this association
of uncertain relevance. Something similar happens with other recently reported clinical
associations with this locus [11]. Joint analysis with our data show only weak
renal disorder and discoid rash due to the differences in effect size between the two
studies. In addition, the other available report shows no association with any of these
three clinical features in any of the two series of patients considered [15], albeit the
analyzed SNP in that report, rs11574637, is only partially correlated with rs1143679.
Of the remaining loci studied here, only TYK2 and BANK1 have previously been
examined for association with SLE clinical features [16-17]. No specific association
was found in the previous studies and we did not find any in ours, in spite of relative
good power. In the interpretation of these negative results, it is important to consider the
power of the each comparison. All the SNPs analyzed here have similar power because
this is determined by the minor allele frequencies that are similar. However, the clinical
features show a gradation of power in function of the rarity of the phenotype with
lowest frequency (Table 2). Larger power is available for clinical features showing a
prevalence approaching 50%, like age of disease onset < 30, photosensitivity or malar
12
130
Procedimiento experimental y resultados
rash, whereas a lower power corresponds to clinical features with low prevalence, like
neurologic disorder, or with very high prevalence, like arthritis or female gender. None
of the latter was associated in our study or has been reported as associated in previous
studies.
Our results and previous reports on the same loci [9-10, 15-17] and in other SLE
susceptibility loci [17, 34-41] do not show a clear and consistent association with
specific clinical features or subphenotypes. Association of the STAT4 locus with some
SLE features is at present the only significant exception to this trend. This conclusion is
not contradicted by some studies that have found differences in SLE susceptibility loci
between patients with a clinical feature and healthy controls, but not differences
between patients with and without the clinical manifestation [16, 39, 41]. Other
confirmed SLE susceptibility loci that have not shown association with specific clinical
manifestations include IRF5 [17, 36-38], PTPN22 [34-35, 39-40] and TNFAIP3 [41].
The same can be affirmed for the MHC, except that in this case, convincing associations
with particular autoantibodies have been reported [42-45]. The available evidence does
not exclude, however, that future studies will find modest or weak associations that
have escaped detection due to lack of power of the current SLE patient collections.
coherent with the fact that they have been identified in studies involving all types of
patients with SLE. It is likely that large phenotype-specific GWAs will uncover genetic
variants with a more clear effect on phenotypes, even if they have less impact in SLE
overall. This type of studies should be actively promoted in the hope that will allow
identifying more uniform subgroups of patients with SLE, which could be very useful to
improve disease management. Also, it is possible that complex interactions between loci
13
131
Factores genéticos asociados con predisposición a LES
phenotypes is supported by ethnic [46-47], ancestry [48] and family studies [49-51], and
it should be expected that future research will be able to identify the phenotype-specific
loci.
CONCLUSIONS
The paucity of sound associations with specific clinical features in our study,
together with other reports on the same loci [9-10, 15-17], or on other confirmed SLE
susceptibility loci [17, 34-41] shows that clinical variability of SLE does not correlate in
a consistent and clear way with SLE susceptibility alleles. The best of the already
known associations is with the STAT4 locus, and it is still incompletely defined. Other
associations that are only tentative at present will be confirmed in new studies, but it is
likely that only phenotype-specific GWAS will lead to the discovery of a significant
ABBREVIATIONS
COMPETING INTEREST
14
132
Procedimiento experimental y resultados
AUTHORS’ CONTRIBUTIONS
MS-G participated in design of the study, genotyped the samples, participated in the
statistical analysis and in the interpretation of results. ML, JO-R, EB, MB, C-GK, CP,
IK-R, PC, F-NS, MM, R-ES, TW, EE, AK, MM, RS, SM, GD-S, M-JS, FV, AS, CG,
IR, F-JB, NB, SD, RP Jr, RP, SR, CD, and JJG-R participated in the acquisition of
clinical data, collection of samples and in the analysis and interpretation of results. AG
participated in the design of the study, coordinated acquisition of clinical data and
results and writing of the manuscript. All authors read and approved the final
manuscript.
ACKNOWLEDGEMENTS
Education. The present work was supported by Fondo de Investigacion Sanitaria of the
Instituto de Salud Carlos III (Spain), grants 06/0620, 08/0744 and RD08/0075/0019 that
are partially financed by the Fondo Europeo de Desarrollo Regional program of the
European Union, by grants from the Xunta de Galicia, and by BMBF KN Rheuma grant
REFERENCES
1. Wallace DJ, Hahn B, Dubois EL: Dubois' lupus erythematosus, 7th edn.
15
133
Factores genéticos asociados con predisposición a LES
2009, 32(3-4):223-230.
14(1):2-12.
5. Karassa FB, Trikalinos TA, Ioannidis JP: The Fc gamma RIIIA-F158 allele is
6. Sestak AL, Nath SK, Sawalha AH, Harley JB: Current status of lupus
7. Moser KL, Kelly JA, Lessard CJ, Harley JB: Recent insights into the genetic
8. Rhodes B, Vyse TJ: The genetics of SLE: an update in the light of genome-
9. Taylor KE, Remmers EF, Lee AT, Ortmann WA, Plenge RM, Tian C, Chung
SA, Nititham J, Hom G, Kao AH, Demirci FY, Kamboh MI, Petri M, Manzi S,
Kastner DL, Seldin MF, Gregersen PK, Behrens TW, Criswell LA: Specificity
16
134
Procedimiento experimental y resultados
dsDNA and shows additive effects with two risk alleles of IRF5. Hum Mol
11. Kim-Howard X, Maiti AK, Anaya JM, Bruner GR, Brown E, Merrill JT, Edberg
JC, Petri MA, Reveille JD, Ramsey-Goldman R, Alarcon GS, Vyse TJ, Gilkeson
G, Kimberly RP, James JA, Guthridge JM, Harley JB, Nath SK: ITGAM
coding variant (rs1143679) influences the risk of renal disease, discoid rash,
14. Dupont WD, Plummer WD, Jr.: Power and sample size calculations. A review
15. Hom G, Graham RR, Modrek B, Taylor KE, Ortmann W, Garnier S, Lee AT,
Chung SA, Ferreira RC, Pant PV, Ballinger DG, Kosoy R, Demirci FY,
17
135
Factores genéticos asociados con predisposición a LES
358(9):900-909.
16. Guo L, Deshmukh H, Lu R, Vidal GS, Kelly JA, Kaufman KM, Dominguez N,
Klein W, Kim-Howard X, Bruner GR, Scofield RH, Moser KL, Gaffney PM,
Dozmorov IM, Gilkeson GS, Wakeland EK, Li QZ, Langefeld CD, Marion MC,
Williams AH, Divers J, Alarcon GS, Brown EE, Kimberly RP, Edberg JC,
10(5):531-538.
18
136
Procedimiento experimental y resultados
19. Pons-Estel BA, Catoggio LJ, Cardiel MH, Soriano ER, Gentiletti S, Villa AR,
21. Petri M: The effect of race on the presentation and course of SLE in the
22. Wang F, Wang CL, Tan CT, Manivasagar M: Systemic lupus erythematosus
6(3):248-253.
24. Hulley SB: Designing clinical research, 3rd edn. Philadelphia, PA: Lippincott
25. Guillemin F: Primer: the fallacy of subgroup analysis. Nat Clin Pract
26. Brookes ST, Whitley E, Peters TJ, Mulheran PA, Egger M, Davey Smith G:
19
137
Factores genéticos asociados con predisposición a LES
28. Harley JB, Alarcon-Riquelme ME, Criswell LA, Jacob CO, Kimberly RP,
Moser KL, Tsao BP, Vyse TJ, Langefeld CD, Nath SK, Guthridge JM, Cobb
BL, Mirel DB, Marion MC, Williams AH, Divers J, Wang W, Frank SG,
Namjou B, Gabriel SB, Lee AT, Gregersen PK, Behrens TW, Taylor KE,
B, Lamas JR, de la Serna AR, Ortiz AM, Carreno L, Canete JD, Caliz R, Blanco
30. Gregersen PK, Amos CI, Lee AT, Lu Y, Remmers EF, Kastner DL, Seldin MF,
Criswell LA, Plenge RM, Holers VM, Mikuls TR, Sokka T, Moreland LW,
Bridges SL, Jr., Xie G, Begovich AB, Siminovitch KA: REL, encoding a
defined risk locus for rheumatoid arthritis. Nat Genet 2009, 41(7):820-823.
CP, Rodriguez L, Vicente E, Gonzalez-Gay MA, Mayes MD, Tan FK, Martin J,
20
138
Procedimiento experimental y resultados
32. Dymecki SM, Niederhuber JE, Desiderio SV: Specific expression of a tyrosine
35. Kyogoku C, Langefeld CD, Ortmann WA, Lee A, Selby S, Carlton VE, Chang
Rodine P, Graham RR, Ardlie KG, Gaffney PM, Moser KL, Petri M, Begovich
21
139
Factores genéticos asociados con predisposición a LES
strong risk factor for systemic lupus erythematosus. Hum Mol Genet 2008,
17(6):872-881.
GD, Blanco FJ, Rego I, Pullmann R, Jr., Pullmann R, Kallenberg CG, Bijl M,
41. Bates JS, Lessard CJ, Leon JM, Nguyen T, Battiest LJ, Rodgers J, Kaufman
KM, James JA, Gilkeson GS, Kelly JA, Humphrey MB, Harley JB, Gray-
22
140
Procedimiento experimental y resultados
43. Galeazzi M, Sebastiani GD, Tincani A, Piette JC, Allegri F, Morozzi G, Bellisai
patients and their relation to clinical features. Ann N Y Acad Sci 2009,
1173:575-580.
45. Fernando MM, Stevens CR, Walsh EC, De Jager PL, Goyette P, Plenge RM,
Vyse TJ, Rioux JD: Defining the role of the MHC in autoimmunity: a review
46. Uribe AG, McGwin G, Jr., Reveille JD, Alarcon GS: What have we learned
vs. nurture) cohort? Where are we heading? Autoimmun Rev 2004, 3(4):321-
329.
23
141
Factores genéticos asociados con predisposición a LES
47. Peschken CA, Katz SJ, Silverman E, Pope JE, Fortin PR, Pineau C, Smith CD,
48. Chung SA, Tian C, Taylor KE, Lee AT, Ortmann WA, Hom G, Graham RR,
BP, Harley JB, Gaffney PM, Moser KL, Manzi S, Petri M, Gregersen PK,
Langefeld CD, Behrens TW, Seldin MF, Criswell LA: European population
2009, 60(8):2448-2456.
49. Ramos PS, Kelly JA, Gray-McGuire C, Bruner GR, Leiran AN, Meyer CM,
Namjou B, Espe KJ, Ortmann WA, Reichlin M, Langefeld CD, James JA,
Gaffney PM, Behrens TW, Harley JB, Moser KL: Familial aggregation and
50. Scofield RH, Bruner GR, Kelly JA, Kilpatrick J, Bacino D, Nath SK, Harley JB:
erythematosus and reveals genetic linkages at 1q22 and 11p13. Blood 2003,
101(3):992-997.
51. Tsao BP, Grossman JM, Riemekasten G, Strong N, Kalsi J, Wallace DJ, Chen
CJ, Lau CS, Ginzler EM, Goldstein R, Kalunian KC, Harley JB, Arnett FC,
24
142
Procedimiento experimental y resultados
25
143
Factores genéticos asociados con predisposición a LES
Table 1: SLE susceptibility loci included in the study with the representative SNPs,
their Minor Allele Frequencies (MAF) in patients and controls, O.R. and P valuesa
b
MAF % (n/N)
c
Locus SNP SLE patients Controls O.R. (95% C.I.) P-value
-15
ITGAM rs1143679 23.0 (718/3116) 15.1 (521/3448) 1.71 (1.5-2.0) 8.1 x 10
-13
STAT4 rs7574865 32.6 (1015/3118) 23.4 (808/3456) 1.53 (1.4-1.7) 6.0 x 10
-6
C8orf13-BLK rs13277113 30.9 (963/3116) 25.2 (868/3444) 1.32 (1.2-1.5) 2.0 x 10
-5
IRAK1-MECP2 rs17435 27.0 (841/3120) 21.5 (743/3456) 1.27 (1.1-1.4) 2.5 x 10
a
These analyses were reported in detail in Suarez-Gestal et al (22)
b
n = number of minor alleles, N = total number of alleles
c
Comparisons were done with logistic regression following a genetic additive model and including
gender as covariate except for IRAK1-MECP2, which is in the X chromosome
26
144
Procedimiento experimental y resultados
Table 2: Clinical characteristics of the patients with SLE included in the study and the
number of subjects with available information for each of them
Number of
Characteristic Value subjects
Women (%) 90.2 1544
a
Age of disease onset (median, IQR ) 29 (21-40) 1340
Age of disease onset < 30 (%) 52.0 1340
b
Time of follow-up (median, IQR) 10 (6-17) 1159
b
Malar rash (%) 56.6 1296
Discoid rash (%) 18.1 1379
Photosensitivity (%) 53.6 1388
Oral ulcers (%) 28.1 1390
Arthritis (%) 80.6 1398
Serositis (%) 35.8 1392
Renal disorder (%) 41.0 1387
Neurologic disorder (%) 14.5 1305
Hematologic disorder (%) 74.2 1348
Immunologic disorder (%) 77.6 1383
b
Antinuclear Antibody (%) 98.5 1129
a
IQR = Inter-quartile range
b
Data from specific recruitment centres were excluded because the feature was underreported: the Czech
Republic and Madrid for time of follow-up; Hungary for malar rash; and the Evangelismos Hospital in
Greece and the Milan collections for ANA
27
145
Factores genéticos asociados con predisposición a LES
Table 3: Comparison of the allele frequencies at nine SNP from SLE susceptibility loci
between patients positive and negative for 11 clinical features. Only significant
differences and results corresponding to previously reported associations are shown.
MAF % (n/N) a
O.R.
Locus Feature Positive patients Negative patients P Pcorr
(95% C.I.)
a
MAF = Minor Allele Frequency, n = number of minor alleles, N = total number of alleles
b
n.s. = not significant
28
146
Procedimiento experimental y resultados
1.32 1.84
C8orf13-BLK Renal disorder 0.0014 0.023
(1.1-1.6) (1.1-3.1)
1.22 2.15
IRAK1-MECP2 Renal disorder 0.029 0.05
(1.0-1.5) (1.0-4.6)
1.15 1.62
STAT4 Age of onset <30 0.075 0.097
(1.0-1.3) (0.9-2.9)
0.78 0.79
BANK1 Oral ulcers 0.018 0.5
(0.6-1.0) (0.4-1.6)
0.79 1.03
Renal disorder 0.018 0.9
(0.7-1.0) (0.6-1.8)
0.79 1.13
PXK Oral ulcers 0.024 0.71
(0.6-1.0) (0.6-2.2)
1.22 1.19
ITGAM Age of onset <30 0.04 0.55
(1.0-1.5) (0.7-2.1)
29
147
Factores genéticos asociados con predisposición a LES
Table 5. Combined analysis of data from current and previous studies. Only
associations significant in any of the studies and allowing for case-only analysis were
considered.
b
OR this OR previous O.R.comb. b c
Locus Feature a Pcomb. Pheter. N
study study (95% C.I.)
1.37 0.73
C8orf13-BLK Renal disorder - - 0.0018 2165
(1.2-1.6) (0.5-1.1)
a
Previous studies were [15] for C8orf13-BLK, [9] for STAT4 and [11] for ITGAM
b
Fixed-effect metaanalysis odds ratio (95 % confidence interval) and P value
c
Pheter = heterogeneity P value; N = total number of patients in the combined analysis
d
Random-effect metaanalysis gave non-significant results
30
148
Procedimiento experimental y resultados
149
Procedimiento experimental y resultados
Population Subgroups
Marchini4, Sergio Migliaresi5, Attila Kovacs6, Josep Ordi Ros7, Marc Bijl8, Maria Jose Santos9,
D’Alfonso14, Gian Domenico Sebastiani15, Ana Suarez16, Francisco J Blanco17, Juan J Gomez-
Reino1,18, Antonio Gonzalez1, and the European Consortium of SLE DNA Collections
ACADEMIC DEGREES:
M Suarez-Gestal, MSc; M Calaza, MSc; T Witte, MD; C Papasteriades, MD; M Marchini, MD; S
Migliaresi, MD; A Kovacs, MD; J Ordi Ros, MD; M Bijl, MD; MJ Santos, MD; S Ruzickova, MD;
R Pullmann, MD; P Carreira, MD; FN Skopouli, MD; S D’Alfonso, MD; GD Sebastiani, MD; A
AFFILIATIONS:
1
Laboratorio de Investigacion 10 and Rheumatology Unit, Instituto de Investigacion Sanitaria -
Greece
151
Factores genéticos asociados con predisposición a LES
4
Clinical Immunology, University of Milan and Fondazione IRCCS Ospedale Maggiore
Barcelona. Spain
8
Department of Rheumatology and Clinical Immunology, University Medical Center Groningen,
Republic
11
Institute of Clinical Biochemistry, Martin Faculty Hospital, Jessenius Medical Faculty, Kollárova
Corresponding author:
Antonio Gonzalez
152
Procedimiento experimental y resultados
Laboratorio de Investigacion 10
15706-Santiago de Compostela
Spain
FUNDING
MS-G is the recipient of a FPU pre-doctoral bursary of the Spanish Ministry of Education. This
work was supported by Fondo de Investigacion Sanitaria of the Instituto de Salud Carlos III
(Spain), grants [06/0620, 08/0744 and by RETICS Program, RD08/0075 (RIER)] that are partially
financed by the Fondo Europeo de Desarrollo Regional program of the European Union, and by
grants from the Xunta de Galicia and by BMBF KN Rheuma grant [C2.12 to TW].
CONFLICT OF INTERESTS
ABBREVIATIONS
153
Factores genéticos asociados con predisposición a LES
154
Procedimiento experimental y resultados
ABSTRACT
We aimed to determine if Systemic Lupus Erythematosus (SLE) phenotypes differ between the two
subgroups of Europeans identified in large genetic studies. Prevalence of ACR SLE classification
criteria and mean age of disease onset were compared between 475 patients with SLE from the
Northern subgroup and 1080 patients from the Southern subgroup. Arthritis was more prevalent in
the Southern subgroup (P = 1.7 x 10-6). Photosensitivity was more prevalent in the Northern group
(P = 0.0006). These differences were concordant with a previous study that analyzed European
ancestry percentages in American patients with SLE, confirming a new level of phenotype
heterogeneity of SLE inside one of the continental ethnic groups and suggesting the need of new
155
Factores genéticos asociados con predisposición a LES
INTRODUCTION
SLE clinical heterogeneity poses many challenges to clinical diagnosis, treatment and
research 11. Genetic, environmental and socioeconomic factors seem to have a role, but our
understanding of their effects is still very incomplete. Variation on severity and clinical phenotype
between ethnic groups has been shown in multiple studies 1, 4, 6, 8, 11-12. Part of this variation is
attributed to differences in the genetic component. A very interesting addition that strengthens the
link between genetic factors and SLE clinical heterogeneity and extends the distinction to
differences within a single ethnic group has been a recent report by Chung et al.3. In this study,
distinction between Northern and Southern European ancestries, which are the 2 major European
subgroups according to genome wide studies 7, correlated with the prevalence of specific SLE
features. Replication of these findings is necessary given the high variability of SLE clinical data
between patient collections that makes this type of studies prone to irreproducibility 1-2, 5-6, 10, 12. It is
also necessary given the important consequences for SLE research in Europeans that will need to
consider separately the 2 subpopulations. Here, we have replicated 2 of the associations described
by Chung et al: arthritis was more frequent in patients from the South of Europe and
METHODS
Patient data: Data from 1555 European SLE patients recruited at 16 centres from 9 different
countries were analyzed 9. They included the American College of Rheumatology (ACR)
classification criteria, age of onset, time of follow-up and gender. All patients and controls gave
their written informed consent and the study was approved by the relevant ethic committees.
Statistical analysis: Patients were divided in Northern and Southern subgroups according with their
place of origin. We excluded from analysis the variables showing large variability within the 2
subgroups (Coefficient of variation (CV) ≥ 30 % between the centres in the Northern or Southern
156
Procedimiento experimental y resultados
Founding sources: This work was supported by Fondo de Investigacion Sanitaria of the Instituto
de Salud Carlos III, the Fondo Europeo de Desarrollo Regional program of the European Union,
grants from the Xunta de Galicia and by BMBF KN Rheuma grant. This founding bodies had no
contribution in designing the study, collecting, analyzing and interpreting the data and approving
RESULTS
The Northern European subgroup was made of 475 patients with SLE from Germany, The
Netherlands, Hungary, the Czech Republic and Slovakia. The Southern Europeans subgroup was
The first step in the analysis was to determine the variability of SLE clinical data within
each of the 2 subgroups. This analysis was done separately for the centres corresponding to
Southern and Northern European subpopulations (Table 1). Discoid rash, oral ulcers, renal disorder
and neurologic disorder showed CV that were larger than 30 % in the Southern and Northern
subgroups and were excluded from further analysis. Serositis, immunologic disorder, malar rash and
antibodies CV were below 20 % (Table 1). Variability in patient gender and age of disease onset
was low. Time of follow-up was more variable in the Southern subgroup and significantly different
between the 2 subgroups (Table 2). However, this difference in time of follow-up had not
appreciable effect in the results except for serositis, which was less prevalent in the patients with
Only arthritis, photosensitivity and serositis were different between the patients from the
Southern and Northern European subpopulations (Table 2). The differences in prevalence of
arthritis and photosensitivity remained clearly significant after accounting for the number of test
157
Factores genéticos asociados con predisposición a LES
performed (Pcorr = 0.000017 and 0.0056, respectively), but not the difference in serositis (Pcorr =
0.054). In addition, the association with serositis disappeared when the difference in follow-up
between the Northern and Southern subpopulations was taken into account (P = 0.6).
Notably, the differences in arthritis and photosensitivity were in the same direction
previously reported in the European Americans 3, with a prevalence in excess of 10.7 % in arthritis
among the patients from the South and of 9.4 % in photosensitivity among the subjects from the
Northern group.
DISCUSSION
Differences in the SLE phenotype between continental ethnic populations are widely
acknowledged 1, 4, 6, 8, 11-12. However, there is only a report indicating that there are also differences
between patients of European origin depending on their Northern or Southern ancestry 3. Here, we
have addressed this question and found 2 clear differences, in arthritis and photosensitivity, that are
concordant with the study of Chung et al. 3. This concordance is very striking because design of the
2 studies has been very different. The Chung et al. study included almost exclusively European
polymorphisms. In contrast, we have used data from European patients living in Europe and their
ancestry has been ascertained by their place of origin. This difference is important because each
collection of patients is affected by environmental exposures and other local factors that could
confound analysis, but these factors will be very different in the 2 studies. Therefore, we find very
reassuring that the arthritis and photosensitivity results of the 2 studies were concordant.
elucidation will help understand SLE clinical heterogeneity. For example, photosensitivity excess in
the Northern subpopulation could be related with a lighter skin colour 3, 8, which is genetically
determined.
Other associations of the Chung et al. study were not replicated in our analysis 3. We did not
158
Procedimiento experimental y resultados
found any difference between the 2 subgroups of Europeans in the prevalence of immunologic
disorder and we did not compare discoid rash or renal disorder because these 2 features showed
large variability within each of the subgroups in our data. High variability in the prevalence clinical
features of SLE is very common between different collections of patients 1-2, 5-6, 10, 12. When it is
In summary, our study replicates the association of the Northern European subpopulation
with more photosensitivity and less arthritis than the observed in the Southern subpopulation.
Concordance between our study and Chung et al. 3 identify SLE clinical features under genetic
influence, advance in our understanding of SLE clinical heterogeneity, and call our attention to the
ACKNOWLEDGMENTS:
Other contributors to the European Consortium of SLE DNA Collections: Myriam Liz,
Scorza, Clinical Immunology, University of Milan and Fondazione IRCCS Ospedale Maggiore
Policlinico, Mangiagalli e Regina Elena, 20122 Milan, Italy; Emöke Endereffy, Department of
Paediatrics, Albert Szent-Györgyi Clinical Centre, University of Szeged, 6720 Hungary; Eva
Balada, Internal Medicine, Research Laboratory in Autoimmune Diseases Hospital Vall d’Hebron,
Immunology, University Medical Center Groningen, 9713 Groningen, The Netherlands; Filipe
159
Factores genéticos asociados con predisposición a LES
Universidade de Lisboa, Portugal; Ctibor Dostal, Molecular Biology and Immunogenetics Dept,
Institute of Rheumatology, 128 50 Prague 2, Czech Republic; Rudolf Pullmann Jr, Gerontology
Research Center, National Institute on Aging, 5600 Nathan Shock Drive, Baltimore, Maryland
21224, USA; Maria Mavromati, Pathophysiology Department, Athens University Medical School,
Athens 115 27, Greece; Nadia Barizzone, Dept Medical Sciences and IRCAD , Eastern Piedmont
University, 28100 Novara, Italy; Carmen Gutierrez, Department of Functional Biology, Hospital
Universitario Central de Asturias, Universidad de Oviedo, Oviedo 33006, Spain; Ignacio Rego,
10
160
Procedimiento experimental y resultados
REFERENCES
1. Alarcon GS, McGwin G, Jr., Petri M, Reveille JD, Ramsey-Goldman R, Kimberly RP.
Cosyns JP, Danieli MG, D DC, Depresseux G, Direskeneli H, Domenech I, Espinosa G, Fernandez-
Nebro A, Ferrara GB, Font J, Frutos MA, Galeazzi M, Garcia-Carrasco M, Garcia Iglesias MF,
Veronesi J, Vianna J, Vivancos J. Systemic lupus erythematosus in Europe at the change of the
millennium: lessons from the "Euro-Lupus Project". Autoimmun Rev 2006;5: 180-6.
3. Chung SA, Tian C, Taylor KE, Lee AT, Ortmann WA, Hom G, Graham RR, Nititham J,
Kelly JA, Morrisey J, Wu H, Yin H, Alarcon-Riquelme ME, Tsao BP, Harley JB, Gaffney PM,
Moser KL, Manzi S, Petri M, Gregersen PK, Langefeld CD, Behrens TW, Seldin MF, Criswell LA.
4. Fernandez M, Alarcon GS, Calvo-Alen J, Andrade R, McGwin G, Jr., Vila LM, Reveille JD.
A multiethnic, multicenter cohort of patients with systemic lupus erythematosus (SLE) as a model
for the study of ethnic disparities in SLE. Arthritis Rheum 2007;57: 576-84.
5. Hom G, Graham RR, Modrek B, Taylor KE, Ortmann W, Garnier S, Lee AT, Chung SA,
11
161
Factores genéticos asociados con predisposición a LES
Ferreira RC, Pant PV, Ballinger DG, Kosoy R, Demirci FY, Kamboh MI, Kao AH, Tian C,
Syvanen AC, Criswell LA, Gregersen PK, Behrens TW. Association of systemic lupus
6. Petri M. The effect of race on the presentation and course of SLE in the United States.
7. Seldin MF, Shigeta R, Villoslada P, Selmi C, Tuomilehto J, Silva G, Belmont JW, Klareskog
8. Sestak AL, Nath SK, Kelly JA, Bruner GR, James JA, Harley JB. Patients with familial and
sporadic onset SLE have similar clinical profiles but vary profoundly by race. Lupus 2008;17:
1004-9.
Ruzickova S, Jose Santos M, Papasteriades C, Marchini M, Skopouli FN, Suarez A, Blanco FJ,
recently identified systemic lupus erythematosus genetic associations: a case-control study. Arthritis
10. Taylor KE, Remmers EF, Lee AT, Ortmann WA, Plenge RM, Tian C, Chung SA, Nititham J,
Hom G, Kao AH, Demirci FY, Kamboh MI, Petri M, Manzi S, Kastner DL, Seldin MF, Gregersen
PK, Behrens TW, Criswell LA. Specificity of the STAT4 genetic association for severe disease
11. Wallace DJ, Hahn B, Dubois EL. Dubois' lupus erythematosus. 7th edn. Philadelphia:
12. Wang F, Wang CL, Tan CT, Manivasagar M. Systemic lupus erythematosus in Malaysia: a
study of 539 patients and comparison of prevalence and disease expression in different racial and
12
162
Procedimiento experimental y resultados
Table 1: Clinical features of SLE patients and variability of their values within the European
Northern and Southern subgroups.
CV (%)
Value N* Northern † Southern †
Women (%) 90.3 1543 3.1 3.3
age of onset (mean, years) 31.1 1349 6.3 10.6
time of follow-up (mean, years) 11.9 1168 12.6 22.8
Malar rash (%) ‡ 56.5 1305 24.8 19.8
Discoid rash (%) 18.1 1388 48.8 59.2
Photosensitivity (%) 53.4 1397 17.6 23.0
Oral ulcers (%) 28.2 1399 56.0 63.1
Arthritis (%) 80.7 1407 15.8 10.4
Serositis (%) 35.8 1401 9.9 26.2
Renal disorder (%) 41.0 1396 32.5 38.9
Neurologic disorder (%) 14.6 1314 66.4 38.4
Hematologic disorder (%) 74.3 1356 15.0 18.8
Immunologic disorder (%) 77.7 1392 22.1 23.0
Antinuclear antibody (%) ‡ 98.7 1138 1.8 2.1
*
Number of patients used for each analysis
†
Northern subgroup: Germany, the Netherlands, Hungary, the Czech Republic and Slovakia. Southern subgroup: Spain,
Italy, Greece and Portugal.
‡
Some data from all patients of specific recruitment centres were excluded because the feature was underreported:
Hungary for malar rash; and the Evangelismos Hospital in Greece and the Milan collections for antinuclear antibody
13
163
Factores genéticos asociados con predisposición a LES
14
164
Procedimiento experimental y resultados
Table 2: Comparison of clinical features between the Northern and Southern subpopulations of
European patients with SLE
Value
Northern Southern P-value * O.R. (95 % C.I.) †
Women (%) 88.6 91.0 0.14
age of onset (mean, years) 31.9 30.8 0.18
time of follow-up (mean, years) 14.2 10.7 5.3 x 10-14
Malar rash (%) 56.3 55.6 0.17 1.01 (0.9-1.2)
Photosensitivity (%) 59.7 50.3 0.00056 1.49 (1.2-1.9)
Arthritis (%) 73.6 84.3 0.0000017 0.51 (0.4-0.7)
Serositis (%) 39.5 33.8 0.0055 ‡ 1.29 (1.1-1.7)
Hematologic disorder (%) 75.5 73.7 0.12 1.11 (0.9-1.4)
Immunologic disorder (%) 77.2 77.9 0.68 0.95 (0.7-1.2)
Antinuclear antibody (%) 98.6 98.5 0.92 1.06 (0.3-3.3)
*
Comparison done with logistic regression including patient gender as covariate
†
Odds ratios and their 95 % confidence intervals were calculated taking as reference the Southern subpopulation of
patients
‡
This difference did not persist after considering differences in follow-up
15
165
4. Componente genético común a LES y AR
Procedimiento experimental y resultados
Objective. Rheumatoid arthritis (RA) and sys- aim of this study was to determine whether 9 other SLE
temic lupus erythematosus (SLE) share some genetic genetic factors are also implicated in RA susceptibility.
factors such as HLA, PTPN22, STAT4, and 6q23. The Methods. A characteristic single-nucleotide poly-
morphism (SNP) in each of 9 genetic factors, ITGAM
(rs1143679), C8orf13–BLK (rs13277113), TYK2
Supported by the Instituto de Salud Carlos III of the Spanish (rs2304256), 1q25.1 (rs10798269), PXK (rs6445975),
Health Ministry (grants PI06/0620 and PI08/0744) and the Xunta de
Galicia (grant 07PXIB918091PR). The Instituto de Salud Carlos III
KIAA1542 (rs4963128), MECP2 (rs17435), BANK1
Research Network provided support for many of the collaborating (rs17266594), and LY9 (rs509749), was studied in 1,635
research teams (grant RD08/0075). patients with RA and 1,906 control subjects from Spain.
1
Marian Suarez-Gestal, MSc, Manuel Calaza, MSc, Rebeca
Dieguez-Gonzalez, PhD, Eva Perez-Pampin, MD, Antonio Gonzalez, The rs7574865 SNP in STAT4 was also included. Anal-
MD, PhD: Hospital Clinico Universitario de Santiago, Santiago de yses were conducted globally and after stratification by
Compostela, Spain; 2Jose Luis Pablos, MD: Hospital 12 de Octubre, sex and clinical features (anti–cyclic citrullinated pep-
Madrid, Spain; 3Federico Navarro, MD: Hospital Universitario Virgen
Macarena, Seville, Spain; 4Javier Narvaez, MD, PhD: Hospital Uni- tide and rheumatoid factor, shared epitope, rheumatoid
versitario de Bellvitge, Barcelona, Spain; 5Jose Luis Marenco, MD: nodules, radiographic changes, sicca syndrome, and
Hospital Universitario de Valme, Seville, Spain; 6Gabriel Herrero-
Beaumont, MD: Fundacion Jimenez Diaz, Madrid, Spain; 7Benjamin
pneumonitis).
Fernandez-Gutierrez, MD, PhD, José Ramón Lamas, PhD: Hospital Results. No association was observed between RA
Clinico San Carlos, Madrid, Spain; 8Arturo Rodriguez de la Serna, and any of the 9 newly identified SLE genetic factors. A
MD: Hospital Santa Creu i Sant Pau, Barcelona, Spain; 9Ana Maria
Ortiz, MD, PhD: Hospital Universitario de la Princesa, Madrid, Spain; meta-analysis using previous data was consistent with
10
Luis Carreño, MD: Hospital Universitario Gregorio Marañon, Ma- these results. In addition, there were no significant
drid, Spain; 11Juan D. Cañete, MD, PhD: Hospital Clinicico and differences between individuals with and those without
Institut d’Investigacions Biomèdiques August Pi I Sunyer, Barcelona,
Spain; 12Rafael Caliz, MD: Hospital Universitario Virgen de las each of the clinical features analyzed, except the fre-
Nieves, Granada, Spain; 13Francisco J. Blanco, MD: Hospital Univer- quency of the minor allele in the C8orf13–BLK locus that
sitario Juan Canalejo, Coruña, Spain; 14Alejandro Balsa, MD: Hospi- was decreased in patients with sicca syndrome (14.6%
tal La Paz, Madrid, Spain; 15Juan J. Gomez-Reino, MD, PhD: Hospital
Clinico Universitario de Santiago and Universitario de Santiago de versus 22.4% in controls; P ⴝ 0.003).
Compostela, Santiago de Compostela, Spain. Conclusion. None of the 9 recently identified SLE
Dr. Fernandez-Gutierrez has received speaking fees from
Bristol-Myers Squibb (less than $10,000). Dr. Balsa has received
risk factors showed association with RA. Therefore,
consulting fees, speaking fees, and/or honoraria from Abbott (less than common genetic factors affecting the pathogenesis of
$10,000). these 2 disorders seem to be limited, revealing that the
Address correspondence and reprint requests to Antonio
Gonzalez, MD, PhD, Laboratorio de Investigacion 10, Hospital genetic component contributes to the different expres-
Clinico Universitario de Santiago, Travesia de Choupana s/n, 15706 sion of these diseases.
Santiago de Compostela, Spain. E-mail: Antonio.Gonzalez.Martinez.
[email protected].
Submitted for publication March 9, 2009; accepted in revised Rheumatoid arthritis (RA) and systemic lupus
form May 17, 2009. erythematosus (SLE) share a complex etiology encom-
2558
169
Factores genéticos asociados con predisposición a LES
passing genetic, environmental, and stochastic compo- Table 1. Clinical characteristics of the 1,635 patients with rheuma-
nents (1,2). Loss of tolerance to self antigens, which toid arthritis*
leads to stimulation of lymphocytes and other immune Female sex 75.0
cells, release of cytokines, activation of complement, and Age at disease onset, median (interquartile range) years 48 (37–57)
Morning stiffness 96.1
the production of autoantibodies, contributes to the Arthritis in ⱖ3 joint areas 99.7
pathogenesis of both RA and SLE (3,4). RA and SLE Arthritis in hand joints 99.0
also share genetic factors such as those in the HLA, Symmetric arthritis 99.3
Rheumatoid nodules (n ⫽ 1,291) 20.1
PTPN22, STAT4, and 6q23 loci (5–8); however, their Rheumatoid factor (n ⫽ 1,504) 72.7
respective clinical phenotypes are clearly different. RA Erosions (n ⫽ 1,502) 70.4
is characterized by symmetric erosive arthritis of the Sicca syndrome (n ⫽ 903) 14.8
Interstitial pneumonitis (n ⫽ 1,520) 2.8
peripheral joints with pannus formation that is chronic Shared epitope carrier (n ⫽ 594) 54.7
and progressive, which causes a characteristic pattern of Anti–citrullinated protein antibody positive (n ⫽ 655) 66.1
pain, rigidity, and deformities. In contrast, arthritis is * Except where indicated otherwise, values are the percent. When data
only one among the diverse array of clinical manifesta- were not available for all patients, the number of patients for whom
tions of SLE and typically is not erosive, does not have data were available is shown.
a progressive course, and most often is not symmetric
(9). Malar rash, photosensitivity, serositis, nephritis,
peripheral and central nerve system disease, and cyto- larger than the number of such factors in RA. Therefore,
penia are indicative of SLE but not RA. The 2 clinical we examined the role in RA of a representative SNP for
syndromes overlap only in some exceptional patients (a each of 10 recently identified SLE genetic factors:
clinical situation that is referred to as rhupus by many STAT4 (which is already known to be shared by SLE and
investigators). Therefore, the 2 diseases should have RA), ITGAM, C8orf13–BLK, PXK, TYK2, KIAA1542,
differential etiologic factors. Such etiologic factors could 1q25.1, BANK1, MECP2, and LY9 (6). All except LY9
be genetic, with some being specific for SLE and others have consistently been associated with SLE (19). How-
being specific for RA. Alternatively, the differential ever, only the SNP in STAT4 was associated with RA in
factors could be environmental or stochastic. our current comparison of more than 1,600 patients and
A common genetic component of RA, SLE, and 1,900 healthy control subjects. A meta-analysis including
other autoimmune diseases has been hypothesized previous data led to similar results. Therefore, it seems
(10,11). This hypothesis was triggered by the observation that RA does not share most of the new SLE genetic
of an increased concurrence of several autoimmune factors, indicating that the genetic component is a
diseases in members of the same family (12,13) and was significant contributor to the different expression of
strongly reinforced by the discovery of the role of HLA these 2 diseases.
alleles in most autoimmune diseases (14) and, later, by
genetics studies in families showing that linkage to
different autoimmune diseases clustered in overlapping PATIENTS AND METHODS
loci (15). Similar findings were also observed in animal Sample collection. We used DNA samples from 1,635
models of autoimmune diseases (16). More recently, patients with RA and 1,906 healthy control subjects of Spanish
some genetic factors with a shared effect in many of ancestry (281 of the control subjects were previously described
by our group in a study of SLE susceptibility [19]). All patients
these diseases have been identified. The most generally with RA met the 1987 revised American College of Rheuma-
shared is PTPN22, but other examples include STAT4, tology (formerly, the American Rheumatism Association) clas-
the 6q23 locus, and possibly CTLA4 and IRF5 (5– sification criteria (20). The clinical characteristics of the pa-
8,17,18). Now, thanks to genome-wide association stud- tients are shown in Table 1. Patients and control subjects gave
ies, we are beginning to have more comprehensive written informed consent to participate in the study. Sample
collection was approved by the respective ethics committees.
knowledge of the genetic component of these diseases. SNP genotyping. A strongly SLE-associated SNP for
The identification of more genetic factors will allow us to each of 10 recently identified SLE genetic factors was selected
determine whether those that are shared by several (Table 2). The 10 SNPs were amplified in a single polymerase
diseases are the rule or the exception. As a consequence, chain reaction (PCR) (Qiagen Multiplex PCR Kit; Qiagen,
we will make advances in the identification of differen- Chatsworth, CA) with 20 ng of genomic DNA and 0.2 M of
each primer. PCR products were purified by digestion with
tial disease factors. exonuclease I (Epicentre Technologies, Madison, WI) and
At the time when this study was planned, the shrimp alkaline phosphatase (SAP; GE Healthcare, Barcelona,
number of recently identified genetic factors in SLE was Spain). Purified PCR products were included in a single-base
170
Procedimiento experimental y resultados
2560 SUAREZ-GESTAL ET AL
Table 2. Allele frequencies for the 10 SNPs identifying SLE genetic factors*
Minor allele frequency, no. (%)
* SLE ⫽ systemic lupus erythematosus; RA ⫽ rheumatoid arthritis; OR ⫽ odds ratio; 95% CI ⫽ 95%
confidence interval.
† P ⫽ 0.028.
‡ This single-nucleotide polymorphism (SNP), which is in chromosome X, was assessed by analysis of
allelic frequencies in women and by carrier analysis in men.
extension reaction with the SNaPshot Multiplex Kit (Applied 99.9%, and genotypes in the control subjects were in
Biosystems, Foster City, CA) and specific probes. After a Hardy-Weinberg equilibrium.
second purification with SAP (GE Healthcare), samples were
analyzed in the ABI Prism 3130xl Genetic Analyzer (Applied
We observed association of the STAT4 SNP
Biosystems), and genotypes were assigned using GeneMapper rs7574865 with RA (odds ratio [OR] 1.14, 95% confi-
software (Applied Biosystems). (Primer and oligonucleotide dence interval CI 1.0–1.3, P ⫽ 0.028) but no significant
sequences are available from the corresponding author.) differences between patients with RA and control sub-
Statistical analysis. Tests for Hardy-Weinberg equili- jects for the other 8 autosomal SNPs or for the MECP2
brium in control samples were done with Haploview (21), using
a threshold of 0.05, without correction for multiple testing. SNP, which is located in the X chromosome and was
Other statistical analyses were done using a customized version analyzed separately in female and male individuals
of the Statistica 7.0 program (StatSoft, Tulsa, OK). Compari- (Table 2). All genotype comparisons were concordant
son of cases and controls was done with allele frequency data with an additive genetic model and gave results similar
using chi-square tests on 2 ⫻ 2 contingency tables. These to the allele comparisons (results not shown). The power
analyses were also conducted after stratifying the samples by
sex and available clinical data. Results from the stratified of these analyses was enough to detect genetic factors
analyses were subjected to Bonferroni correction. Univariate showing an OR between 1.20 for the ITGAM SNP and
logistic regression models were used to test the fit to the data 1.15 for the LY9 SNP (␣ ⫽ 0.05 and 1 –  ⫽ 0.80), which
of additive, recessive, and dominant genetic models. Statistical were the SNPs with minimum and maximum power,
power was estimated using “power and sample size calcula- respectively.
tions” software (22). Meta-analysis was done with our data and
data from previous reports, using a fixed-effects model with R Analysis was also done after stratification of
software (http://www.r-project.org/). Heterogeneity of the ef- samples according to sex and clinical data, which in-
fect size between studies was assessed with the I2 statistic for cluded anti–cyclic citrullinated peptide antibodies, rheu-
inconsistency and Cochran’s Q statistic. Samples from Hospital matoid factor, the shared epitope, rheumatoid nodules,
Universitario La Paz and Hospital Clinico San Carlos have radiographic changes, sicca symptoms, and interstitial
already been used for the analysis of 2 genetic factors included
in this project: BANK1 and STAT4. To avoid data duplication, pneumonitis. Most of these comparisons showed similar
we excluded these samples from the relevant analysis. results across strata (detailed data are available from the
corresponding author). The most remarkable difference
was the decreased frequency of the minor allele of
RESULTS
rs13277113 in the C8orf13–BLK locus among patients
A total of 1,635 patients with RA and 1,906 with RA and sicca symptoms (14.6% versus 24.2% in
control subjects of Spanish ancestry were available for patients without these symptoms [P ⫽ 0.0005] and versus
study. The genotyping call rate across the 10 SNPs was 22.4% in controls [P ⫽ 0.003]) (Table 3). These differ-
171
Factores genéticos asociados con predisposición a LES
Table 3. Significant differences in stratified analyses of patients with RA, according to sex and clinical features*
Comparisons between patient strata Comparisons with controls
SNP (locus) MAF, no. (%) OR (95% CI) P Pcorr† OR (95% CI) P Pcorr
rs6445975 (PXK)
Women 541/2,252 (24.0) 1.10 (0.9–1.3) NS NS 1.19 (1.0–1.4) 0.015 NS
Men 168/752 (22.3) 0.87 (0.7–1.1) NS NS
rs13277113 (C8orf13–BLK)
Sicca syndrome 39/268 (14.6) 0.53 (0.4–0.8) 0.0005 0.008 0.59 (0.4–0.8) 0.003 0.04
No sicca syndrome 356/1472 (24.2) 1.11 (1.0–1.3) 0.16 NS
rs509749 (LY9)
Shared epitope 330/650 (50.8) 1.41 (1.1–1.8) 0.003 0.05 1.18 (1.0–1.4) 0.05 NS
No shared epitope 227/538 (42.2) 0.84 (0.7–1.0) 0.05 NS
rs10798269 (1q25.1)
RF positive 673/2,184 (30.8) 0.85 (0.7–1.0) 0.06 NS 0.88 (0.8–1.0) 0.02 NS
RF negative 283/822 (34.4) 1.03 (0.9–1.2) NS NS
rs2304256 (TYK2)
Erosions 565/2,108 (26.8) 1.19 (1.0–1.4) 0.06 NS 0.99 (0.9–1.1) NS NS
No erosions 209/888 (23.5) 0.83 (0.7–1.0) 0.04 NS
* The presence of erosions was defined radiographically. RA ⫽ rheumatoid arthritis; SNP ⫽ single-nucleotide polymorphism; MAF ⫽ minor allele
frequency; OR ⫽ odds ratio; 95% CI ⫽ 95% confidence interval; NS ⫽ not significant; RF ⫽ rheumatoid factor.
† Bonferroni correction.
ences remained significant after correction for the num- SNP rs1143679, for which no previous information was
ber of tests performed (Table 3). However, this result found. The meta-analysis confirmed the results observed
should be taken with caution given the small number of in our sample collection: the one SNP that is clearly
patients in this group (n ⫽ 134) and association with a associated with RA is the STAT4 SNP (Table 4), for
different allele than that in patients with SLE. There which ⬎35,000 subjects have already been studied. Re-
were other differences between strata (Table 3), but they sults for this SNP showed significant heterogeneity of
were significant in the analyses only previous to Bonfer- effects between studies (I2 ⫽ 53.0%, P for Cochran’s Q
roni correction. statistic [PQ] ⫽ 0.007) that was not attributable to the
Finally, we combined our results with previous inclusion of our results (I2 ⫽ 53.2%, PQ ⫽ 0.008). This
data including data available only as supplementary heterogeneity prevents a sound estimate of the summary
information and data imputed from the available geno- effect size, but it does not question association of the
types but not directly tested in previous studies. These STAT4 SNP with RA, because all studies showed an OR
analyses included, at least, an additional data set for all of ⬎1.0 for this SNP. Two other SNPs showed border-
of the SNPs except MECP2 SNP rs17435 and ITGAM line association with RA: the SNP in the C8orf13–BLK
Table 4. Combined analysis of allele frequency differences for SLE-associated SNPs obtained in the current and previous studies (including
imputed genotypes) comparing patients with RA and controls*
SNP (locus) No. of RA patients No. of controls OR (95% CI) P Ref.
rs7574865 (STAT4)† 17,565 18,315 1.25 (1.2–1.3) 1.1 ⫻ 10⫺34 31,38–46‡
rs7574865 (STAT4)§ 16,181 16,509 1.26 (1.2–1.3) 2.9 ⫻ 10⫺34 31,38–46‡
rs13277113 (C8orf13–BLK) 3,494 4,562 1.09 (1.0–1.2) 0.03 31
rs2304256 (TYK2) 3,161 2,763 0.94 (0.9–1.0) NS 18
rs10798269 (1q25.1) 5,006 6,694 0.97 (0.9–1.0) NS 31,46
rs4963128 (KIAA1542) 5,014 6,694 1.01 (1.0–1.1) NS 31,46
rs6445975 (PXK) 5,017 6,695 1.06 (1.0–1.1) 0.04 31,46
rs17266594 (BANK1) 3,550 3,960 0.97 (0.9–1.0) NS 37
rs509749 (LY9) 5,014 6,691 1.04 (1.0–1.1) NS 31,46
* SLE ⫽ systemic lupus erythematosus; SNPs ⫽ single-nucleotide polymorphisms; RA ⫽ rheumatoid arthritis; OR ⫽ odds ratio; 95% CI ⫽ 95%
confidence interval; NS ⫽ not significant.
† Analysis of heterogeneity between studies was possible only for STAT4, for which significant heterogeneity was observed (I2 ⫽ 53.0%, P for
Cochran’s Q statistic [PQ] ⫽ 0.007).
‡ Data from refs. 39 and 46 overlap, and only the largest set from ref. 39 was used here.
§ I2 ⫽ 53.2%, PQ ⫽ 0.008, not including data from the present study.
172
Procedimiento experimental y resultados
2562 SUAREZ-GESTAL ET AL
locus for which ⬃8,000 subjects have been analyzed (P ⫽ factors have been independently confirmed [19,35]).
0.03) and the SNP in PXK, with data from ⬎11,000 Therefore, our specific analysis together with the meta-
subjects (P ⫽ 0.04). analysis including previous data allow for a more con-
clusive assessment that reinforces the SLE specificity of
these genetic factors in relation to RA.
DISCUSSION
Our study has enough power to detect effect sizes
During the last decades, evidence supporting a corresponding to an OR between 1.15 and 1.20 for the
common genetic component for a diversity of auto- SNPs in autosomes and 1.24 for the MECP2 SNP in
immune diseases has been continuously investigated chromosome X. This is in the range of the weakest
(10,11,15,16,23). The genetic factors that were associ- detectable effects by analysis of the largest RA sample
ated with a certain disease were considered good candi- collections, such as the North American Rheumatoid
dates for the other diseases in the group, and they were Arthritis Consortium family collection and the Epidemi-
systematically examined. This practice could have intro- ological Investigation of Rheumatoid Arthritis (36).
duced a bias supporting the commonality hypothesis, Combining our results with those from previous studies
because extensive investigation of the same factors in (including imputed data) increased the power. Two
multiple diseases could have led to some associations weak association signals with ORs of 1.09 and 1.06 were
just by chance. This is likely in the context of the lack of detected in the C8orf13–BLK and PXK loci, respectively.
reproducibility that until recently characterized genetics It is therefore possible that SNP effects below these sizes
studies of complex diseases (24). Accordingly, conflict- could have escaped our detection. In this case, the
ing results have been the rule. Nowadays, genetics eventual RA-associated SNPs would be of lower signif-
studies have achieved a high degree of reproducibility icance for RA than for SLE disease, in which the
due to a significant increase in sample size and to more reported ORs for these SNPs have been larger than 1.19,
strict thresholds for claiming association (25). In addi- with the exception of single reports that did not replicate
tion, association across the genome has been analyzed in the LY9 and TYK2 associations (19,35). It is also possible
an unbiased manner (25). These developments gave us that different polymorphisms in the same gene are
the opportunity to address the question of genetic involved in different diseases, as indicated by a recent
commonality between RA and SLE with a new set of report of association of BANK1 SNPs with RA in a study
solidly established genetic factors. that did not show association with the rs17266594 SNP
The results provide evidence against widespread that we examined here (37).
sharing of genetic factors between RA and SLE. Of the Another possibility we have addressed is that the
9 newly examined SLE genetic factors, none was found sharing of genetic factors is restricted to specific sub-
to be associated with RA. This lack of association groups of patients with RA, perhaps with more similarity
persisted after a meta-analysis including previous data, with SLE. This is what happens with polymorphisms in
which makes it unlikely that any of the 9 representative IRF5 that are associated with SLE and with a not-fully-
SNPs have an effect in RA similar to that observed in defined subgroup of patients with RA (17,18). However,
SLE. our analysis did not identify any clear association of this
In retrospect, it could be contended that our type. There were some association signals in the strati-
findings were expected, given the identification of these fied analyses, but only differences in the C8orf13–BLK
genetic factors in the SLE genome-wide association SNP in patients with sicca syndrome remained signifi-
study (26–28) and not in the RA genome-wide associa- cant after correction. Unfortunately, no stratified data
tion study (29–31). However, the lack of significant were available in previous reports that would allow us to
association in a genome-wide association study does not draw more sound conclusions by increasing statistical
mean the lack of any association, because genome-wide power through meta-analysis.
association studies apply strict thresholds for signifi- In summary, the genetic factors in the HLA,
cance, and coverage of the different SLE and RA PTPN22, 6q23, IRF5, and STAT4 loci are shared by SLE,
genome-wide association studies was not identical (i.e., RA, and other immune-mediated diseases (5,6). How-
only 1 of the 10 SNPs studied here was included in the ever, none of the other 9 recently identified SLE genetic
largest RA genome-wide association study [31]). In factors examined here showed association with RA.
addition, 3 SLE genetic factors explored here, TYK2 These results indicate that the genetic component of
(32), MECP2 (33), and LY9 (34), were not found in the each of these 2 diseases contributes significantly to the
SLE genome-wide association study (although the first 2 different disease phenotypes. This is an idea that is not
173
Factores genéticos asociados con predisposición a LES
new but that lacked experimental support (10). The 11. Becker KG. The common variants/multiple disease hypothesis of
common complex genetic disorders. Med Hypotheses 2004;62:
magnitude of the differential component cannot be 309–17.
estimated yet, because a large fraction of the heritability 12. Lin JP, Cash JM, Doyle SZ, Peden S, Kanik K, Amos CI, et al.
of RA and SLE has not been defined. More precise Familial clustering of rheumatoid arthritis with other autoimmune
estimates will be possible once the causal variants of diseases. Hum Genet 1998;103:475–82.
13. Payami H, Joe S, Thomson G. Autoimmune thyroid disease in
each of the genetic factors are identified. It is reasonable type I diabetic families. Genet Epidemiol 1989;6:137–41.
to expect that further genetic research will provide a 14. Fernando MM, Stevens CR, Walsh EC, De Jager PL, Goyette P,
clearer definition of what molecules and pathways are Plenge RM, et al. Defining the role of the MHC in autoimmunity:
a review and pooled analysis. PLoS Genet 2008;4:e1000024.
shared or specific in the pathogenesis of RA and SLE, 15. Becker KG, Simon RM, Bailey-Wilson JE, Freidlin B, Biddison
allowing for better management of these 2 rheumatic WE, McFarland HF, et al. Clustering of non-major histocompat-
diseases. ibility complex susceptibility candidate loci in human autoimmune
diseases. Proc Natl Acad Sci U S A 1998;95:9979–84.
16. Griffiths MM, Encinas JA, Remmers EF, Kuchroo VK, Wilder
RL. Mapping autoimmunity genes. Curr Opin Immunol 1999;11:
ACKNOWLEDGMENT 689–700.
We thank the sample donors for their generous collab- 17. Dieguez-Gonzalez R, Calaza M, Perez-Pampin E, de la Serna AR,
Fernandez-Gutierrez B, Castaneda S, et al. Association of inter-
oration.
feron regulatory factor 5 haplotypes, similar to that found in
systemic lupus erythematosus, in a large subgroup of patients with
rheumatoid arthritis. Arthritis Rheum 2008;58:1264–74.
AUTHOR CONTRIBUTIONS
18. Sigurdsson S, Padyukov L, Kurreeman FA, Liljedahl U, Wiman
All authors were involved in drafting the article or revising it AC, Alfredsson L, et al. Association of a haplotype in the
critically for important intellectual content, and all authors approved promoter region of the interferon regulatory factor 5 gene with
the final version to be published. Dr. Gonzalez had full access to all of rheumatoid arthritis. Arthritis Rheum 2007;56:2202–10.
the data in the study and takes responsibility for the integrity of the 19. Suarez-Gestal M, Calaza M, Endreffy E, Pullmann R, Ordi-Ros J,
data and the accuracy of the data analysis. Domenico Sebastiani G, et al. Replication of recently identified
Study conception and design. Suarez-Gestal, Gonzalez. systemic lupus erythematosus genetic associations: a case control
Acquisition of data. Dieguez-Gonzalez, Perez-Pampin, Pablos, Na- study. Arthritis Res Ther 2009;11:R69.
varro, Narvaez, Marenco, Herrero-Beaumont, Fernandez-Gutierrez, 20. Arnett FC, Edworthy SM, Bloch DA, McShane DJ, Fries JF,
Lamas, Rodriguez de la Serna, Ortiz, Carreño, Cañete, Caliz, Blanco, Cooper NS, et al. The American Rheumatism Association 1987
Balsa, Gomez-Reino, Gonzalez. revised criteria for the classification of rheumatoid arthritis.
Analysis and interpretation of data. Suarez-Gestal, Calaza, Gomez- Arthritis Rheum 1988;31:315–24.
Reino, Gonzalez. 21. Barrett JC, Fry B, Maller J, Daly MJ. Haploview: analysis and
visualization of LD and haplotype maps. Bioinformatics 2005;21:
263–5.
REFERENCES 22. Dupont WD, Plummer WD Jr. Power and sample size calcula-
tions: a review and computer program. Control Clin Trials 1990;
1. Gregersen PK. Genetics of rheumatoid arthritis: confronting 11:116–28.
complexity. Arthritis Res 1999;1:37–44. 23. Raman K, Mohan C. Genetic underpinnings of autoimmunity:
2. Nath SK, Kilpatrick J, Harley JB. Genetics of human systemic lessons from studies in arthritis, diabetes, lupus and multiple
lupus erythematosus: the emerging picture. Curr Opin Immunol sclerosis. Curr Opin Immunol 2003;15:651–9.
2004;16:794–800. 24. Ioannidis JP, Ntzani EE, Trikalinos TA, Contopoulos-Ioannidis
3. Ronnblom L, Pascual V. The innate immune system in SLE: type
DG. Replication validity of genetic association studies. Nat Genet
I interferons and dendritic cells. Lupus 2008;17:394–9.
2001;29:306–9.
4. Malmstrom V, Trollmo C, Klareskog L. The additive role of innate
25. Todd JA. Statistical false positive or true disease pathway? Nat
and adaptive immunity in the development of arthritis. Am J Med
Genet 2006;38:731–3.
Sci 2004;327:196–201.
5. Bowes J, Barton A. Recent advances in the genetics of RA 26. International Consortium for Systemic Lupus Erythematosus Ge-
susceptibility. Rheumatology (Oxford) 2008;47:399–402. netics (SLEGEN), Harley JB, Alarcon-Riquelme ME, Criswell
6. Rhodes B, Vyse TJ. The genetics of SLE: an update in the light of LA, Jacob CO, Kimberly RP, et al. Genome-wide association scan
genome-wide association studies. Rheumatology (Oxford) 2008; in women with systemic lupus erythematosus identifies suscepti-
47:1603–11. bility variants in ITGAM, PXK, KIAA1542 and other loci. Nat
7. Graham RR, Cotsapas C, Davies L, Hackett R, Lessard CJ, Leon Genet 2008;40:204–10.
JM, et al. Genetic variants near TNFAIP3 on 6q23 are associated 27. Kozyrev SV, Abelson AK, Wojcik J, Zaghlool A, Linga Reddy
with systemic lupus erythematosus. Nat Genet 2008;40:1059–61. MV, Sanchez E, et al. Functional variants in the B-cell gene
8. Musone SL, Taylor KE, Lu TT, Nititham J, Ferreira RC, Ortmann BANK1 are associated with systemic lupus erythematosus. Nat
W, et al. Multiple polymorphisms in the TNFAIP3 region are Genet 2008;40:211–6.
independently associated with systemic lupus erythematosus. Nat 28. Hom G, Graham RR, Modrek B, Taylor KE, Ortmann W, Garnier
Genet 2008;40:1062–4. S, et al. Association of systemic lupus erythematosus with C8orf13-
9. Fernandez A, Quintana G, Rondon F, Restrepo JF, Sanchez A, BLK and ITGAM-ITGAX. N Engl J Med 2008;358:900–9.
Matteson EL, et al. Lupus arthropathy: a case series of patients 29. Raychaudhuri S, Remmers EF, Lee AT, Hackett R, Guiducci C,
with rhupus. Clin Rheumatol 2006;25:164–7. Burtt NP, et al. Common variants at CD40 and other loci confer
10. Gregersen PK, Behrens TW. Genetics of autoimmune diseases: risk of rheumatoid arthritis. Nat Genet 2008;40:1216–23.
disorders of immune homeostasis. Nat Rev Genet 2006;7:917–28. 30. Plenge RM, Cotsapas C, Davies L, Price AL, de Bakker PI, Maller
174
Procedimiento experimental y resultados
2564 SUAREZ-GESTAL ET AL
J, et al. Two independent alleles at 6q23 associated with risk of TW, et al. STAT4 and the risk of rheumatoid arthritis and systemic
rheumatoid arthritis. Nat Genet 2007;39:1477–82. lupus erythematosus. N Engl J Med 2007;357:977–86.
31. Wellcome Trust Case Control Consortium. Genome-wide associ- 40. Barton A, Thomson W, Ke X, Eyre S, Hinks A, Bowes J, et al.
ation study of 14,000 cases of seven common diseases and 3,000 Re-evaluation of putative rheumatoid arthritis susceptibility genes
shared controls. Nature 2007;447:661–78. in the post-genome wide association study era and hypothesis of a
32. Sigurdsson S, Nordmark G, Goring HH, Lindroos K, Wiman AC, key pathway underlying susceptibility. Hum Mol Genet 2008;17:
Sturfelt G, et al. Polymorphisms in the tyrosine kinase 2 and 2274–9.
interferon regulatory factor 5 genes are associated with systemic 41. Kobayashi S, Ikari K, Kaneko H, Kochi Y, Yamamoto K, Shimane
lupus erythematosus. Am J Hum Genet 2005;76:528–37. K, et al. Association of STAT4 with susceptibility to rheumatoid
33. Sawalha AH, Webb R, Han S, Kelly JA, Kaufman KM, Kimberly arthritis and systemic lupus erythematosus in the Japanese popu-
RP, et al. Common variants within MECP2 confer risk of systemic lation. Arthritis Rheum 2008;58:1940–6.
42. Orozco G, Alizadeh BZ, Delgado-Vega AM, Gonzalez-Gay MA,
lupus erythematosus. PLoS ONE 2008;3:e1727.
Balsa A, Pascual-Salcedo D, et al. Association of STAT4 with
34. Cunninghame Graham DS, Vyse TJ, Fortin PR, Montpetit A, Cai
rheumatoid arthritis: a replication study in three European popu-
YC, Lim S, et al. Association of LY9 in UK and Canadian SLE
lations. Arthritis Rheum 2008;58:1974–80.
families. Genes Immun 2008;9:93–102.
43. Martinez A, Varade J, Marquez A, Cenit MC, Espino L, Per-
35. Cunninghame Graham DS, Akil M, Vyse TJ. Association of digones N, et al. Association of the STAT4 gene with increased
polymorphisms across the tyrosine kinase gene, TYK2 in UK SLE susceptibility for some immune-mediated diseases. Arthritis
families. Rheumatology (Oxford) 2007;46:927–30. Rheum 2008;58:2598–602.
36. Plenge RM, Padyukov L, Remmers EF, Purcell S, Lee AT, 44. Zervou MI, Sidiropoulos P, Petraki E, Vazgiourakis V, Kra-
Karlson EW, et al. Replication of putative candidate-gene associ- soudaki E, Raptopoulou A, et al. Association of a TRAF1 and a
ations with rheumatoid arthritis in ⬎4,000 samples from North STAT4 gene polymorphism with increased risk for rheumatoid
America and Sweden: association of susceptibility with PTPN22, arthritis in a genetically homogeneous population. Hum Immunol
CTLA4, and PADI4. Am J Hum Genet 2005;77:1044–60. 2008;69:567–71.
37. Orozco G, Abelson AK, Gonzalez-Gay MA, Balsa A, Pascual- 45. Palomino-Morales RJ, Rojas-Villarraga A, Gonzalez CI, Ramirez
Salcedo D, Garcia A, et al. Study of functional variants of the G, Anaya JM, Martin J. STAT4 but not TRAF1/C5 variants
BANK1 gene in rheumatoid arthritis. Arthritis Rheum 2009;60: influence the risk of developing rheumatoid arthritis and systemic
372–9. lupus erythematosus in Colombians. Genes Immun 2008;9:
38. Lee HS, Remmers EF, Le JM, Kastner DL, Bae SC, Gregersen 379–82.
PK. Association of STAT4 with rheumatoid arthritis in the Korean 46. Plenge RM, Seielstad M, Padyukov L, Lee AT, Remmers EF,
population. Mol Med 2007;13:455–60. Ding B, et al. TRAF1-C5 as a risk locus for rheumatoid arthritis:
39. Remmers EF, Plenge RM, Lee AT, Graham RR, Hom G, Behrens a genomewide study. N Engl J Med 2007;357:1199–209.
175
Procedimiento experimental y resultados
Corresponding author:
Antonio Gonzalez
Laboratorio de Investigacion 10
Hospital Clinico Universitario de Santiago
177
Factores genéticos asociados con predisposición a LES
178
Procedimiento experimental y resultados
ABSTRACT
Introduction: Recent studies have shown that TNFAIP3 is associated with several
in SLE, but not in psoriasis. We aimed to complete analysis of this gene in RA.
Methods: Five TNFAIP3 SNPs, including the nsSNP associated with SLE and the SNP
associated with psoriasis, rs610604, and a PERP SLE-associated SNP were analyzed in
1657 patients with RA and 1638 controls of Spanish ancestry. Data were complemented
with genotypes for other SNPs previously studied in the same samples. Stratification of
patients by gender, ACPA, RF, ANA status and by HLA-DRB1 genotypes was done.
Results: Association was observed with rs610604, but not with SLE associated SNPs or
haplotypes. However, association with rs610604 differed from the previously reported
indicated that the rs610604 SNP reflects best the association between RA and
TNFAIP3. ANA status identified differential association of SNPs in the 6q23 region
better than ACPA status, although both were correlated and the associations reinforced
Conclusions: The association of TNFAIP3 with RA was different from the observed in
SLE and in psoriasis. Identification of the involved mechanisms will unravel the
relationships between these autoimmune diseases. In addition, ANA status was the best
179
Factores genéticos asociados con predisposición a LES
INTRODUCTION
Recent research has uncovered several rheumatoid arthritis (RA) genetic factors that are
because it has been identified in a region of chromosome 6q23 devoid of genes and
because it includes variants showing two independent effects [2-4]. These two effects
are identified by the rs6920220 SNP, which minor allele is associated with increased
RA susceptibility, and by the rs10499194 SNP (or the rs13207033 SNP), which minor
associations, we and others have found evidence for a combined effect of these two
SNPs in the intergenic locus with SNPs in the tumour necrosis factor-Į-induced protein
multiple levels and, therefore, a good candidate for an important role in RA. In addition,
both has been also observed in systemic lupus erythematosus (SLE) [7-9], psoriasis
[10], celiac disease [11], type 1 diabetes [12] and juvenile idiopatic arthritis [13-14],
indicating that genetic variation in this broad 6q23 region influences susceptibility to
previous analysis [5], which was done with tagSNPs, the most informative SNPs from
new reports in other diseases and by analysing specific subphenotypes that could be
shared between them. The SLE studies have been the most exhaustive and rewarding
because they have found strong association with SNPs at the two loci, the intergenic
180
Procedimiento experimental y resultados
region and the TNFAIP3 gene, and with a third locus: the PERP gene that is further 207
Kb downstream from TNFAIP3 [7-9]. In addition, these SLE studies showed strong
association with a TNFAIP3 nsSNP (rs2230926 or its proxy in our samples, rs5029939)
[7-9] that was shown to hinder the inhibitory effect of TNFAIP3 on NFkappaB [8].
These results make of this nsSNP the most likely TNFAIP3 causal variant in SLE
susceptibility and suggests a potential mechanism of action. In addition, our new study
has included other SNPs because psoriasis shows no association with any of the SNPs
that are associated with SLE in the intergenic region or in the TNFAIP3 locus, but with
follow-up study of RA the most associated SNP in PERP, rs6922466, and five
TNFAIP3 SNPs: the nsSNP rs2230926 and two other SNPs strongly associated to SLE
rs7749323 and rs5029939, the rs610604 SNP associated to psoriasis and a tagSNP,
patients with differential association to any of these SNPs. The six SNPs were analyzed
in 1657 patients with RA and 1638 controls. Results showed association with the
psoriasis susceptibility SNP in TNFAIP3, but the direction of change was the opposite.
in this region, but these associations did not match with the described in SLE. These
results suggest the possibility that the causal variants in this locus may be different in
181
Factores genéticos asociados con predisposición a LES
DNA samples: Samples used in this study (1657 patients with RA and 1638 controls)
have already been described [5]. All patients met the revised American College of
Rheumatology 1987 classification criteria [15]. Participants gave their written informed
Genotyping: Six SNPs were genotyped in multiplex PCR and single-base extension
reactions as described [5]. PCR primers and hybridization probes are detailed in
using the AlleleSEQR HLA-DRB1 Typing kit (Abbott, Germany), which includes a
single PCR amplification for all the alleles of the second exon of DRB1 and
the frequency of one of the allele combinations over the alternative in our Spanish
sample.
Statistical analysis: Quality control, comparison of allele and genotype frequency data,
conditional logistic regression, haplotype analysis and statistical power estimation were
carried out as described [5]. Data from our previous study on this region was included
RESULTS
The five TNFAIP3 SNPs and one PERP SNP were genotyped with a 99.9 % call rate
allele frequencies between the 1657 patients with RA and the 1638 controls showed
weak significant differences only in two SNPs: rs610604 and rs644340 (O.R. = 0.89
182
Procedimiento experimental y resultados
and 0.90, respectively; Table 1) that are, respectively, the SNP associated with psoriasis
and the tagSNP included to complete coverage of the locus. Both showed a modest
protective effect of their minor allele. No association was observed with any of the other
three TNFAIP3 SNPs that are the most associated in SLE studies (Table 1). As these
three SLE susceptibility SNPs have minor allele frequencies below 5 %, the power of
these analyses could be a matter of concern. However, it was possible to exclude with
great confidence a similar effect than the reported in SLE (power > 99.9 % to detect an
effect of O.R. = 2.0 with p < 0.001). This is consequence of the large size of our study
and the considerable magnitude of the effects reported in SLE (O.R. 2.0)[7-9]. In
addition, the PERP rs6922466 SNP did not show allele differences between patients
with RA and controls in our study. Again, power of our study was enough (> 90 % for p
< 0.001) to detect an effect as large as the reported in SLE (O.R. = 0.77)[8]. Genotype
Integration of these and our previous results showed that rs610604 was the TNFAIP3
SNP most strongly associated with RA. However, it was in tight LD with other two
associated SNPs, rs582757 and rs644340, and their individual effects were redundant by
conditional logistic regression analysis (not shown). That is, association with any of the
three SNPs disappeared when any of the other two was included in a multilocus model.
These results suggest that there is a single causal polymorphism in TNFAIP3 for the RA
association.
Analysis of TNFAIP3 haplotypes with the ten SNPs (five from our previous report, and
five from this study) showed no difference between cases and controls (Table 2). In
contrast, the SLE associated haplotype defined by the rare allele of rs7749323 was not
associated with RA. We also combined the enlarged data from the TNFAIP3 locus with
our results from the 6q23 intergenic locus [5]. As could be expected by the redundancy
183
Factores genéticos asociados con predisposición a LES
between rs582757, rs610604 and rs644340, any of these three SNPs was present in the
best regression logistic models together with SNPs of the intergenic locus (not shown).
These models showed association with RA in both the 6q23 intergenic region and in the
from the newly studied SNPs in TNFAIP3 or in PERP was found. Therefore, none of
the analyses performed, single SNP, haplotype or multilocus, have provided new
Given the discrepant associations between SLE and RA in the TNFAIP3 locus we were
with SLE-like characteristics (clinical features are shown in supplementary table 2). In
our previous analyses we stratified by gender, ACPA and RF status and we detected
associations between women and ACPA positive patients and SNPs of the intergenic
locus, but none with SNPs of the TNFAIP3 locus. Similarly, none of the new SNPs was
differentially associated when stratified by these three clinical variables (not shown). In
addition, we have analyzed all the SNPs after stratification by the presence of ANA, the
shared epitope and SLE-associated HLA-DRB1 alleles (*0301, *1501 and *0801).
regarding association with the intergenic and the TNFAIP3 loci. There were 5 SNPs
with different frequencies between ANA positive and ANA negative patients (Table 3).
In addition, 4 SNPs were different between ANA positive patients and controls, and 2
between ANA negative patients and controls. Surprisingly, none of the three TNFAIP3
SNPs associated with SLE, rs2230926, rs7749323 and rs5029939, were significantly
the 4 SNPs different in ANA positive patients were explained by rs13207033, which
showed the most marked difference with controls. This SNP was associated only with
184
Procedimiento experimental y resultados
the RA patients that were ANA positive in our collection of samples (Table 3). On the
other side, the two SNPs that were specially associated with the ANA negative
subgroup of patients were redundant, although rs610604 explained better the association
in conditional analysis than rs11970361 (not shown). This SNP, which explained all
association in the TNFAIP3 locus, was only associated in the ANA negative subgroup,
with no difference between the ANA positive patients and the controls (Table 3). None
of the stratifications on HLA alleles identified any differential association in the two
The marked effect of ANA status motivated us to test if the ANA positive patients were
different from the ANA negative ones in the presence of ACPA, RF, SE alleles and SLE
associated HLA-DRB1 alleles (Table 4). The comparisons showed that ANA positive
patients were similar to the ANA negative patients in all these features except in the
presence of RF that was more common among the ANA positive subjects. Therefore,
there was not any indication to suspect of the classification as RA of the ANA positive
patients.
Our previous analyses have showed that association of rs13207033 was restricted to
ACPA positive patients. Therefore, we checked how the two subgroups of antibody
positive patients were related. Most ANA positive patients were also ACPA positive
(66.2 %, Table 4), but association was not different in the ANA positive patients that
were ACPA negative and in the positive for the two autoantibodies (O.R. = 0.67 vs
0.74, respectively). Therefore, the ANA status had a more marked effect in association
with this SNP than the ACPA status and was able to explain it entirely.
DISCUSION
Our follow-up study has not identified any new independent association with RA in the
TNFAIP3 locus or in the PERP gene. This indicates that although association in this
185
Factores genéticos asociados con predisposición a LES
region of chromosome 6q23 is complex, it could be tractable. In addition, our study has
helped to clarify it by showing that the pattern of association with RA in TNFAIP3 does
not resemble SLE or psoriasis. The most associated SNP was rs610604, as described in
psoriasis, but the risk alleles were the opposite. The rare allele has been described as
associated with increased risk in psoriasis, but it was associated with protection in RA.
In addition, the SLE susceptibility SNPs were not associated with RA. This lack of
association of the SLE SNPs was observed in spite of a very good power for a similar
effect. Also psoriasis has been found to lack association with the SLE SNPs in
TNFAIP3. This pattern is important because suggests that association of the TNFAIP3
locus with several diseases cannot be assumed to be due to the same polymorphism and
mechanisms. However, these results require replication in independent studies given the
weak association in the locus we have found and lack of concordance with the results of
two recent studies that showed association of RA with the SLE-associated rs2230926,
weak in a UK study (P = 0.03, O.R. = 1.21)[6] and stronger in the Japanese (P = 2.6 x
because these diseases share multiple characteristics, but they also have distinctive
features and their identification can be very useful to understand the possibilities and
the case of RA and SLE, there is clear evidence of a set of loci that are shared. They
include PTPN22, STAT4, the 6q23 intergenic locus, C8orf13–BLK and PXK, and,
probably, IRF5 and CTLA4 [17-18]. Other associations seem to be specific either of
10
186
Procedimiento experimental y resultados
SLE. As mentioned, our results suggest that association with TNFAIP3 will be of this
type too, present in the two diseases but with different alleles. The genetic relationship
between RA and psoriasis is less clear. They do not share any of the clearly
demonstrated susceptibility loci, not even PTPN22. An apparent exception could have
been association with the TNFAIP3 locus (psoriasis does not show association with
SNPs in the 6q23 intergenic locus) [10], but the same variant showed opposed effects in
Heterogeneity in the association with the 6q23 region has also been observed within the
patients with RA since the first reports [2-6, 16]. It seems that one of the two
rs10499194, is predominant in patients with ACPA [3, 5, 16]. Now, we have found that
it was also more marked in patients with ANA. In fact, the presence of ANA was able to
account for the effect of ACPA status in this association in spite of identifying a much
smaller fraction of the patients. In addition, ANA status was differentially associated
with SNPs in the TNFAIP3 locus, but not with the SLE-associated SNPs in this locus, as
could have been expected. These results are intriguing because they suggest that the
add to the evidence of heterogeneous effects of genetic variation in the 6q23 region.
However, these resutls should be interpreted with caution because not all reports are
concordant in the relationship between ACPA subphenotypes and SNPs in the region
[6], and because there were limitations in our stratification on ANA. These limitations
ANA data for a fraction of the patients and assessment of ANA status at different times
in the evolution of RA. Specifically, some of the ANA data were obtained after
11
187
Factores genéticos asociados con predisposición a LES
classification of the ANA positive patients as genuine RA: apart from being classified
CONCLUSIONS
In summary, this follow-up study indicates that only three association signals in the
6q23 region contribute to RA susceptibility: two in the intergenic locus and one in
PERP was excluded. Results also suggest that the association in TNFAIP3 is different
from the described in SLE and in psoriasis, although this result will require
confirmation. Our study also highlights the possibility that ANA status could be useful
ABBREVIATIONS
COMPETING INTEREST
12
188
Procedimiento experimental y resultados
AUTHORS’ CONTRIBUTION
MS-G participated in design of the study, genotyped the samples, participated in the
genotyping. GH-B, J-DC, A-MO, J-LM, RC, BF-G, JN, AR-S, LC, AB, F-JB, EP-P, J-
LP and FN participated in the acquisition of clinical data and collection of samples and
in the analysis and interpretation of results. JJG-R coordinated the acquisition of clinical
data and collection of samples and participated in the analysis and interpretation of
results. AG participated in the design of the study and in the coordination of acquisition
interpretation of results and writing of the manuscript. All authors read and approved
ACKNOWLEDGMENTS
We thank Carmen Pena-Pena for her excellent technical assistance. M.S-G is the
the recipient of an “Isabel Barreto” bursary of the Government of Galicia. This project
was supported by grants PI080744 and by RETICS Program, RD08/0075 (RIER) from
the Instituto de Salud Carlos III (Spain) with participation of funds from FEDER
(European Union).
REFERENCES
13
189
Factores genéticos asociados con predisposición a LES
14
190
Procedimiento experimental y resultados
12. Fung EY, Smyth DJ, Howson JM, Cooper JD, Walker NM, Stevens H, Wicker
LS, Todd JA: Analysis of 17 autoimmune disease-associated variants in type
1 diabetes identifies 6q23/TNFAIP3 as a susceptibility locus. Genes Immun
2009, 10(2):188-191.
13. Prahalad S, Hansen S, Whiting A, Guthery SL, Clifford B, McNally B, Zeft AS,
Bohnsack JF, Jorde LB: Variants in TNFAIP3, STAT4, and C12orf30 loci
associated with multiple autoimmune diseases are also associated with
juvenile idiopathic arthritis. Arthritis Rheum 2009, 60(7):2124-2130.
14. Hinks A, Eyre S, Ke X, Barton A, Martin P, Flynn E, Packham J, Worthington J,
Thomson W: Overlap of disease susceptibility loci for rheumatoid arthritis
(RA) and juvenile idiopathic arthritis (JIA). Ann Rheum Dis 2009.
15. Arnett FC, Edworthy SM, Bloch DA, McShane DJ, Fries JF, Cooper NS, Healey
LA, Kaplan SR, Liang MH, Luthra HS, et al.: The American Rheumatism
Association 1987 revised criteria for the classification of rheumatoid
arthritis. Arthritis Rheum 1988, 31(3):315-324.
16. Shimane K, Kochi Y, Horita T, Ikari K, Amano H, Hirakata M, Okamoto A,
Yamada R, Myouzen K, Suzuki A, Kubo M, Atsumi T, Koike T, Takasaki Y,
Momohara S, Yamanaka H, Nakamura Y, Yamamoto K: The association of a
nonsynonymous single-nucleotide polymorphism in TNFAIP3 with systemic
lupus erythematosus and rheumatoid arthritis in the Japanese population.
Arthritis Rheum 2010, 62(2):574-579.
17. Suarez-Gestal M, Calaza M, Dieguez-Gonzalez R, Perez-Pampin E, Pablos JL,
Navarro F, Narvaez J, Marenco JL, Herrero-Beaumont G, Fernandez-Gutierrez
B, Lamas JR, de la Serna AR, Ortiz AM, Carreno L, Canete JD, Caliz R, Blanco
FJ, Balsa A, Gomez-Reino JJ, Gonzalez A: Rheumatoid arthritis does not
share most of the newly identified systemic lupus erythematosus genetic
factors. Arthritis Rheum 2009, 60(9):2558-2564.
18. Gregersen PK, Amos CI, Lee AT, Lu Y, Remmers EF, Kastner DL, Seldin MF,
Criswell LA, Plenge RM, Holers VM, Mikuls TR, Sokka T, Moreland LW,
Bridges SL, Jr., Xie G, Begovich AB, Siminovitch KA: REL, encoding a
member of the NF-kappaB family of transcription factors, is a newly
defined risk locus for rheumatoid arthritis. Nat Genet 2009, 41(7):820-823.
19. Charles PJ, Smeenk RJ, De Jong J, Feldmann M, Maini RN: Assessment of
antibodies to double-stranded DNA induced in rheumatoid arthritis
patients following treatment with infliximab, a monoclonal antibody to
tumor necrosis factor alpha: findings in open-label and randomized
placebo-controlled trials. Arthritis Rheum 2000, 43(11):2383-2390.
15
191
Factores genéticos asociados con predisposición a LES
Table 1. Comparison of the minor allele frequencies of the five TNAFIP3 and the PERP
a
MAF % (n/N)
SNP RA patients Controls O.R. (95% C.I.) b P
TNFAIP3
PERP
a
MAF = Minor Allele Frequency, n = number of minor alleles, N = total number of alleles
b
O.R. = Odds Ratio, C.I. = Confidence Interval
c
ns = not significant
16
192
Procedimiento experimental y resultados
and controls
a
Corresponding SNPs: 1: rs600144; 2: rs11970361; 3: rs11970411; 4: rs5029939; 5: rs2230926; 6:
17
193
194
Table 3. Analysis of the allele frequencies in the 6q23 intergenic and TNFAIP3 loci after stratification of patients with RA by ANA status
a + -
MAF % (n/N) ANA vs. controls ANA vs. controls
+ - b + -
SNP ANA ANA Controls O.R. (95% C.I.) P O.R. (95% C.I.) P P ANA vs ANA
rs13207033 23.0 (100/434) 28.2 (305/1082) 29.7 (914/3082) 0.71 (0.6-0.9) 0.0044 0.93 (0.8-1.1) 0.36 0.041
rs6920220 24.4 (110/450) 21.2 (240/1134) 20.1 (639/3184) 1.29 (1.0-1.6) 0.032 1.07 (0.9-1.3) 0.43 0.16
rs694069 33.4 (149/446) 38.7 (436/1126) 40.0 (1276/3188) 0.75 (0.6-0.9) 0.0073 0.95 (0.8-1.1) 0.44 0.05
rs600144 20.9 (95/454) 27.7 (329/1188) 26.8 (857/3202) 0.72 (0.6-0.9) 0.008 1.05 (0.9-1.2) 0.54 0.0051
rs11970361 8.3 (38/458) 4.3 (51/1192) 6.0 (194/3242) 1.42 (1.0-2.0) 0.056 0.70 (0.5-1.0) 0.028 0.0012
rs610604 35.8 (164/458) 30.3 (361/1190) 34.3 (1122/3272) 1.07 (0.9-1.3) 0.52 0.83 (0.7-1.0) 0.013 0.033
Factores genéticos asociados con predisposición a LES
a
MAF = Minor Allele Frequency, n = number of minor alleles, N = total number of alleles
b
O.R. = Odds Ratio, C.I. = Confidence Interval
Procedimiento experimental y resultados
a
ANA % (n/N)
positive negative P
a
n = number of subjects with the characteristic; N = total number of available subjects with complete
information
195
196
Supplementary Table 1. Primers and probes used for genotyping the TNFAIP3 and PERP SNPs represented in the 5’ to 3’ direction
PCR primers
Locus SNP Forward Primer Reverse Primer Minisequencing Probe
TNFAIP3 rs5029939 ccctgtgtgctcctccttag tgaatgacaccaactgcaaag TTACCTATGAacaccaactgcaaaggagccatgggaccttggttctagcttaa $
TNFAIP3 rs2230926 ttgctgggtcttacatgcag cccgtttcaacaaattcctg
aggacacagacttggtactgaggaaggcgctgt
TNFAIP3 rs610604 tggatggaccaagattgatt tgtggattatggcaagaattgta TTACCTATGATTGATCGTGGTGATATCCGattgtagaaccattcactgaaatgcaggcctgtgatgaagagct
$
TNFAIP3 rs644340 tagctggaattacaggcgctc aggaggctgaggtgcgcag TTACCTATGATTGATgaggctgaggtgcgcagatcacttaaggccaagagtcaaaaac $
TNFAIP3 rs7749323 gtctgagccttgtccctgag tgggtctcctctcttgtgct ggtagaccatggaatccacattaagaca
PERP rs6922466 gtctgttccgccaccattac acagctgaattcaaaagtgga ttaattgtatcattcagttttgtctggggaagtttttgggttttgttt
Primers were selected with Primer3 and Oligos. They were checked to avoid formation of dimers between the primers included in the reaction.
Factores genéticos asociados con predisposición a LES
$ These oligonucleotides were extended with a 5' tail that has not homology with human sequences (these tails are in capital letters)
Procedimiento experimental y resultados
197
DISCUSIÓN
Discusión
1. Discusión
201
Factores genéticos asociados con predisposición a LES
202
Discusión
variables clínicas, aunque éstas fueron débiles. Por otra parte, se trató de buscar
si existía una correlación entre las manifestaciones clínicas y la ascendencia
noreuropea o sureuropea de los pacientes. Los resultados confirmaron una
mayor frecuencia de fotosensibilidad en el norte y de artritis en el sur,
sugiriendo una influencia modesta del fondo genético de estas dos
subpoblaciones de europeos en la variabilidad clínica del LES.
203
Factores genéticos asociados con predisposición a LES
204
Discusión
205
Factores genéticos asociados con predisposición a LES
206
Discusión
207
Factores genéticos asociados con predisposición a LES
más fuerte que los de MECP2 (OR > 1.5) (Gateva V y col., 2009; Jacob CO y
col., 2009). Así pues, es importante que estudios futuros traten de determinar
cuál de estos dos genes es el responsable del efecto causal. Después de esto,
todavía quedaría identificar el SNP funcional.
208
Discusión
209
Factores genéticos asociados con predisposición a LES
210
Discusión
codifica para una integrina que se une a una amplia variedad de ligandos (Ehlers
MR y col., 2000). Esta molécula media la adhesión leucocitaria, la
trasmigración a través del endotelio y la fagocitosis de moléculas opsonizadas
con C3bi. Esta última función es importante para la eliminación de células
apoptóticas por los macrófagos (Mevorach D y col., 1998), proceso defectuoso
en pacientes con LES (Ren Y y col., 2003) que, por tanto, podría contribuir a la
patogenia de la enfermedad (Casciola-Rosen LA y col., 1994).
211
Factores genéticos asociados con predisposición a LES
Por último, el SNP rs13277113 (OR = 1.34) está localizado entre dos
genes que se transcriben en direcciones opuestas, C8orf13 y BLK. Este
polimorfismo se había descrito en el GWAS de Hom y col., con un efecto
similar al encontrado (OR = 1.39) (Hom G y col., 2008). Otros dos estudios
encontraron asociaciones significativas en este locus, uno de ellos con un SNP
en BLK fuertemente ligado a rs13277113 (Graham RR y col., 2008), y el otro
con un SNP independiente (SLEGEN y col., 2008). Estos resultados sugieren la
posibilidad de la existencia de dos efectos genéticos independientes en la región.
El locus también se ha visto asociado recientemente con AR (Gregersen PK y
col., 2009) y esclerosis sistémica (Gourh P y col., 2010). No se ha identificado
ningún polimorfismo que tuviese un efecto funcional. Aún así, se observó que
el alelo de riesgo de rs13277113 se correlaciona con niveles bajos de mRNA de
BLK y altos de C8orf13. Por lo tanto, cualquiera de estos dos efectos podría
tener un papel en el LES. Sin embargo, mientras que C8orf13 es un gen de
función desconocida, BLK es un buen candidato en el LES porque está
implicado en la ruta de señalización del BCR y en la diferenciación de células
pro-B a pre-B (Tretter T y col., 2003). Además, como se comentó anteriormente,
existen evidencias de que BLK interacciona con otro factor de susceptibilidad al
212
Discusión
Uno de los factores genéticos de mayor interés en el LES hasta hace poco
fue PDCD1, dado su papel crítico como inmunoregulador en múltiples
enfermedades y su localización en un locus de ligamiento (Prokunina L y col.,
2002; Keir ME y col., 2007; Day CL y col., 2006; Urbani S y col., 2006). El
SNP PD1.3, localizado en el cuarto intrón de este gen, fue propuesto como el
polimorfismo causal responsable de la asociación con LES (Prokunina L y col.,
2002). Esto se debe a que se encuentra en una región con características
compatibles con un enhancer. La región presenta múltiples sitios de unión a
factores de transcripción relacionados con funciones inmunes y 4 repeticiones
en tandem de unos 40 nucleótidos. La única evidencia experimental de que
PD1.3 puede repercutir funcionalmente sobre el gen PDCD1 era un EMSA. En
este ensayo se veía alterada la unión del factor de transcripción RUNX1,
relacionado con la diferenciación hematopoyética (Prokunina L y col., 2002).
213
Factores genéticos asociados con predisposición a LES
214
Discusión
215
Factores genéticos asociados con predisposición a LES
216
Discusión
esto no excluiría una posible relevancia funcional, ya que una fracción de los
elementos funcionales no se han conservado. Otra característica de los enhancer
típicos es que se encuentran en posición 5’ ó 3’ al gen o en el primer intrón del
mismo, no habiéndose descrito en otros intrones. Así pues, estas observaciones
junto con los resultados obtenidos en los ensayos de gen reporter y en los
EMSAs cuestionan la relevancia funcional de este sitio en la regulación de la
expresión de PDCD1 y en la susceptibilidad al LES. Al excluirse PD1.3, queda
abierta la posibilidad de que otro polimorfismo de PDCD1 fuese el responsable
de la susceptibilidad. Sin embargo, no hay ningún candidato claro y los GWAS
de LES publicados recientemente no mostraron ninguna señal de asociación en
este locus, por lo que en la actualidad no está confirmado que PDCD1 sea un
factor de susceptibilidad al LES.
217
Factores genéticos asociados con predisposición a LES
En primer lugar, los 9 loci asociados con LES que se han estudiado
(ITGAM, KIAA1542, PXK, 1q25.1, C8orf13-BLK, STAT4, TYK2, MECP2 y
BANK1) se analizaron estratificando por las manifestaciones clínicas (10
fenotipos definidos por los criterios de clasificación de la ACR y edad de
aparición de la enfermedad). Las asociaciones observadas fueron débiles y poco
consistentes. La asociación más clara fue la del alelo menor del SNP
rs13277113 de C8orf13-BLK con mayor riesgo de nefritis. Por otra parte,
STAT4 es el factor genético que ha mostrado asociaciones más sólidas con
variables clínicas en varios estudios. Sin embargo, el alelo menor del SNP
rs7574865 de STAT4 sólo mostró asociación con edad temprana de la
enfermedad y una tendencia de asociación con menor susceptibilidad a úlceras
orales. La escasez de asociaciones consistentes se debe, en parte, a las
limitaciones de este tipo de análisis.
218
Discusión
219
Factores genéticos asociados con predisposición a LES
220
Discusión
Una segunda asociación, aunque menos clara, fue la del SNP rs17435 de
IRAK1-MECP2 con nefritis. Se observó tanto en el análisis global, como de
forma consistente en mujeres y hombres. Sin embargo, esta asociación no
persistió tras realizar la corrección por múltiples tests. Por lo tanto, es necesario
confirmarla en estudios futuros.
221
Factores genéticos asociados con predisposición a LES
222
Discusión
223
Factores genéticos asociados con predisposición a LES
224
Discusión
225
Factores genéticos asociados con predisposición a LES
226
Discusión
De los 9 loci asociados con LES que se han estudiado (STAT4, ITGAM,
C8orf13-BLK, TYK2, 1q.25.1, PXK, KIAA1542, MECP2 y BANK1), sólo STAT4
había sido ampliamente analizado y claramente asociado con AR (Wellcome
Trust Case Control Consortium, 2007; Lee HS y col., 2007; Remmers EF y col.,
2007; Barton A y col., 2008; Kobayashi S y col., 2008; Orozco G y col., 2008;
Martinez A y col., 2008; Zervou MI y col., 2008b; Palomino-Morales RJ y col.,
2008). También se confirmó este resultado en nuestra colección de muestras de
AR. Sin embargo, no se encontró ninguna asociación significativa para los otros
8 factores genéticos, a pesar de que el poder estadístico de este estudio era
suficiente para detectar efectos con OR de 1.15-1.20 de SNPs en cromosomas
autosómicos y 1.24 del SNP de MECP2, en el cromosoma X. Además, estos
efectos serían similares a las OR más débiles encontradas en las dos grandes
colecciones de muestras de AR, NARAC (North American Rheumatoid
Arthritis Consortium) y EIRA (Swedish Epidemiological Investigation of
Rheumatoid Arthritis) (Plenge RM y col., 2005).
227
Factores genéticos asociados con predisposición a LES
Así pues, parece poco probable que estos SNPs tengan una importancia similar
en ambas enfermedades.
Algo similar parece ocurrir con TNFAIP3. Por un lado, los SNPs de baja
frecuencia, rs5029939, rs2230926 y rs7749323, fuertemente asociados con LES
(OR ≥ 2, P ≤ 10-4) (Musone SL y col., 2008; Graham RR y col., 2008), no
mostraron asociación con AR. Por otro lado, el SNP rs610604 presentó una
asociación con AR cuyo efecto era opuesto al observado en psoriasis (Nair RP y
col., 2009). Sin embargo, dicho SNP junto con el rs644340 mostraban un efecto
protector similar al del SNP que ya se había estudiado con anterioridad
(rs582757) (Dieguez-Gonzalez R y col., 2009) y con la que tienen cierta
relación. Por tanto, se confirmó dicha señal protectora en el gen TNFAIP3.
Como ya se había mostrado previamente, ésta contribuye a la susceptibilidad a
AR de forma conjunta con las señales protectora (rs13207033, rs10499194) y
de riesgo (rs6920220) localizadas en la región intergénica de 6q23. Así pues, el
patrón de asociación con AR en TNFAIP3 parece distinto al de LES o psoriasis.
Sin embargo, estos resultados en AR necesitan ser confirmados en estudios
228
Discusión
229
Factores genéticos asociados con predisposición a LES
230
Discusión
2. Sumario
231
Factores genéticos asociados con predisposición a LES
232
Discusión
Por último, observamos que una buena parte de los nuevos factores
genéticos de LES eran específicos de LES, ya que no estaban asociados con AR.
Así, el único factor de susceptibilidad de los 9 analizados que está claramente
compartido por ambas enfermedades es STAT4. C8orf13-BLK y PXK mostraron
una débil asociación con AR, pero sólo al combinar nuestros datos con los de
otros estudios. En cuanto a TNFAIP3, nuestros resultados muestran señales de
asociación distintas en las dos enfermedades. En resumen, los factores genéticos
que son compartidos por el LES y la AR son PTPN22, STAT4, la región
intergénica de 6q23 y puede que IRF5, CTLA4, C8orf13-BLK, PXK y BANK1.
Sin embargo, el componente genético de ambas enfermedades difiere en
muchos otros factores de susceptibilidad, como TNFAIP3, KIAA1542, 1q25.1,
MECP2 y TYK2. Estos resultados indican que, aunque comparten una fracción
233
Factores genéticos asociados con predisposición a LES
234
CONCLUSIONES
Conclusiones
Conclusiones
237
Factores genéticos asociados con predisposición a LES
238
BIBLIOGRAFÍA
Bibliografía
Bibliografía
241
Factores genéticos asociados con predisposición a LES
242
Bibliografía
susceptibility candidate loci in human autoimmune diseases. Proc Natl Acad Sci
U S A. 1998; 95: 9979-9984.
• Becker KG. The common variants/multiple disease hypothesis of common
complex genetic disorders. Med Hypotheses. 2004; 62: 309-317.
• Begovich AB, Carlton VE, Honigberg LA, Schrodi SJ, Chokkalingam AP,
Alexander HC et al. A missense single-nucleotide polymorphism in a gene
encoding a protein tyrosine phosphatase (PTPN22) is associated with
rheumatoid arthritis. Am J Hum Genet. 2004; 75: 330-337.
• Bellizzi D, Rose G, Cavalcante P, Covello G, Dato S, De Rango F et al. A
novel VNTR enhancer within the SIRT3 gene, a human homologue of SIR2, is
associated with survival at oldest ages. Genomics. 2005; 85: 258-263.
• Beyaert R, Heyninck K, Van Huffel S. A20 and A20-binding proteins as
cellular inhibitors of nuclear factor-kappa B-dependent gene expression and
apoptosis. Biochem Pharmacol. 2000; 60: 1143-1151.
• Blank C, Brown I, Marks R, Nishimura H, Honjo T, Gajewski TF et al.
Absence of programmed death receptor 1 alters thymic development and
enhances generation of CD4/CD8 double-negative TCR-transgenic T cells. J
Immunol. 2003; 171: 4574-4581.
• Boh EE. Neonatal lupus erythematosus. Clin Dermatol. 2004; 22: 125-128.
• Boone DL, Turer EE, Lee EG, Ahmad RC, Wheeler MT, Tsui C et al. The
ubiquitin-modifying enzyme A20 is required for termination of Toll-like
receptor responses. Nat Immunol. 2004; 5: 1052-1060.
• Bottini N, Musumeci L, Alonso A, Rahmouni S, Nika K, Rostamkhani M et
al. A functional variant of lymphoid tyrosine phosphatase is associated with
type I diabetes. Nat Genet. 2004; 36: 337-338.
• Botto M, Walport MJ. C1q, autoimmunity and apoptosis. Immunobiology.
2002; 205: 395-406.
243
Factores genéticos asociados con predisposición a LES
244
Bibliografía
the millennium: lessons from the "Euro-Lupus Project". Autoimmun Rev. 2006;
5: 180-186.
• Chahrour M, Jung SY, Shaw C, Zhou X, Wong ST, Qin J et al. MeCP2, a key
contributor to neurological disease, activates and represses transcription.
Science. 2008; 320: 1224-1229.
• Chang YK, Yang W, Zhao M, Mok CC, Chan TM, Wong RWS et al.
Association of BANK1 and TNFSF4 with systemic lupus erythematosus in Hong
Kong Chinese. Genes Immun. 2009; 10: 414-420.
• Charles PJ, Smeenk RJ, De Jong J, Feldmann M, Maini RN. Assessment of
antibodies to double-stranded DNA induced in rheumatoid arthritis patients
following treatment with infliximab, a monoclonal antibody to tumor necrosis
factor alpha: findings in open-label and randomized placebo-controlled trials.
Arthritis Rheum. 2000; 43: 2383-2390.
• Chowdhury D, Beresford PJ, Zhu P, Zhang D, Sung JS, Demple B et al. The
exonuclease TREX1 is in the SET complex and acts in concert with NM23-H1 to
degrade DNA during granzyme A-mediated cell death. Mol Cell. 2006; 23: 133-
142.
• Chung SA, Criswell LA. PTPN22: its role in SLE and autoimmunity.
Autoimmunity. 2007; 40: 582-590.
• Chung SA, Tian C, Taylor KE, Lee AT, Ortmann WA, Hom G et al.
European population substructure is associated with mucocutaneous
manifestations and autoantibody production in systemic lupus erythematosus.
Arthritis Rheum. 2009; 60: 2448-2456.
• Cooper GS, Gilbert KM, Greidinger EL, James JA, Pfau JC, Reinlib L et al.
Recent advances and opportunities in research oln Lupus: environmental
influences and mechanisms of disease. Environ Health Perspect. 2008; 116:
695-702.
245
Factores genéticos asociados con predisposición a LES
• Coxon A, Rieu P, Barkalow FJ, Askari S, Sharpe AH, von Andrian UH et al.
A novel role for the beta 2 integrin CD11b/CD18 in neutrophil apoptosis: a
homeostatic mechanism in inflammation. Immunity. 1996; 5: 653-666.
• Coyle AJ, Gutierrez-Ramos JC. The expanding B7 superfamily: increasing
complexity in costimulatory signals regulating T cell function. Nat Immunol.
2001; 2: 203-209.
• Crotty S, Johnston RJ, Schoenberger SP. Effectors and memories: Bcl-6 and
Blimp-1 in T and B lymphocyte differentiation. Nat Immunol. 2010; 11: 114-120.
• Cunninghame Graham DS, Akil M, Vyse TJ. Association of polymorphisms
across the tyrosine kinase gene, TYK2 in UK SLE families. Rheumatology
(Oxford). 2007a; 46: 927-930.
• Cunninghame Graham DS, Graham RR, Manku H, Wong AK, Whittaker JC,
Gaffney PM et al. Polymorphism at the TNF superfamily gene TNFSF4 confers
susceptibility to systemic lupus erythematosus. Nat Genet. 2008a; 40: 83-89.
• Cunninghame Graham DS, Manku H, Wagner S, Reid J, Timms K, Gutin A
et al. Association of IRF5 in UK SLE families identifies a variant involved in
polyadenylation. Hum Mol Genet. 2007b; 16: 579-591.
• Cunninghame Graham DS, Vyse TJ, Fortin PR, Montpetit A, Cai YC, Lim S
et al. Association of LY9 in UK and Canadian SLE families. Genes Immun.
2008b; 9:93-102.
• Day CL, Kaufmann DE, Kiepiela P, Brown JA, Moodley ES, Reddy S et al.
PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion
and disease progression. Nature. 2006; 443: 350-354.
• Deapen D, Escalante A, Weinrib L, Horwitz D, Bachman B, Roy-Burman P
et al. A revised estimate of twin concordance in systemic lupus erythematosus.
Arthritis Rheum. 1992; 35: 311-318.
246
Bibliografía
247
Factores genéticos asociados con predisposición a LES
248
Bibliografía
• Finnegan A, Grusby MJ, Kaplan CD, O'Neill SK, Eibel H, Koreny T et al. IL-
4 and IL-12 regulate proteoglycan-induced arthritis through Stat-dependent
mechanisms. J Immunol. 2002; 169: 3345-3352.
• Forabosco P, Gorman JD, Cleveland C, Kelly JA, Fisher SA, Ortmann WA et
al. Meta-analysis of genome-wide linkage studies of systemic lupus
erythematosus. Genes Immun. 2006; 7: 609-614.
• Frank R, Zhang J, Uchida H, Meyers S, Hiebert SW, Nimer SD et al. The
AML1/ETO fusion protein blocks transactivation of the GM-CSF promoter by
AML1B. Oncogene. 1995; 11: 2667-2674.
• Franke A, Balschun T, Karlsen TH, Sventoraityte J, Nikolaus S, Mayr G et al.
Sequence variants in IL10, ARPC2 and multiple other loci contribute to
ulcerative colitis susceptibility. Nat Genet. 2008; 40: 1319-1323.
• Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H et al.
Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family
member leads to negative regulation of lymphocyte activation. J Exp Med. 2000;
192: 1027-1034.
• Fung EY, Smyth DJ, Howson JM, Cooper JD, Walker NM, Stevens H et al.
Analysis of 17 autoimmune disease-associated variants in type 1 diabetes
identifies 6q23/TNFAIP3 as a susceptibility locus. Genes Immun. 2009; 10:
188-191.
• Gaffney PM, Kearns GM, Shark KB, Ortmann WA, Selby SA, Malmgren ML
et al. A genome-wide search for susceptibility genes in human systemic lupus
erythematosus sibpair families. Proc Natl Acad Sci U S A. 1998; 95: 14875-
14879.
• Gaffney PM, Ortmann WA, Selby SA, Shark KB, Ockenden TC, Rohlf KE et
al. Genome screening in human systemic lupus erythematosus: results from a
second Minnesota cohort and combined analyses of 187 sib-pair families. Am J
Hum Genet. 2000; 66: 547-556.
249
Factores genéticos asociados con predisposición a LES
250
Bibliografía
251
Factores genéticos asociados con predisposición a LES
252
Bibliografía
253
Factores genéticos asociados con predisposición a LES
254
Bibliografía
• Jones PL, Veenstra GJ, Wade PA, Vermaak D, Kass SU, Landsberger N et al.
Methylated DNA and MeCP2 recruit histone deacetylase to repress
transcription. Nat Genet. 1998; 19: 187-191.
• Kamachi Y, Ogawa E, Asano M, Ishida S, Murakami Y, Satake M et al.
Purification of a mouse nuclear factor that binds to both the A and B cores of
the polyomavirus enhancer. J Virol. 1990; 64: 4808-4819.
• Karassa FB, Trikalinos TA, Ioannidis JP. The Fc gamma RIIIA-F158 allele is
a risk factor for the development of lupus nephritis: a meta-analysis. Kidney Int.
2003; 63: 1475-1482.
• Kariuki SN, Kirou KA, MacDermott EJ, Barillas-Arias L, Crow MK,
Niewold TB. Cutting edge: autoimmune disease risk variant of STAT4 confers
increased sensitivity to IFN-alpha in lupus patients in vivo. J Immunol. 2009;
182: 34-38.
• Kassel R, Cruise MW, Iezzoni JC, Taylor NA, Pruett TL, Hahn YS.
Chronically inflamed livers up-regulate expression of inhibitory B7 family
members. Hepatology. 2009; 50: 1625-1637.
• Kaufman KM, Kelly JA, Herring BJ, Adler AJ, Glenn SB, Namjou B et al.
Evaluation of the Genetic Association of the PTPN22 R620W Polymorphism in
Familial and Sporadic Systemic Lupus Erythematosus. Arthritis Rheum. 2006;
54: 2533-2540.
• Kawasaki A, Ito I, Hikami K, Ohashi J, Hayashi T, Goto D et al. Role of
STAT4 polymorphisms in systemic lupus erythematosus in a Japanese
population: a case-control association study of the STAT1-STAT4 region.
Arthritis Res Ther. 2008a; 10: R113.
• Kawasaki A, Kyogoku C, Ohashi J, Miyashita R, Hikami K, Kusaoi M et al.
Association of IRF5 polymorphisms with systemic lupus erythematosus in a
Japanese population: support for a crucial role of intron 1 polymorphisms.
Arthritis Rheum. 2008b; 58: 826-834.
255
Factores genéticos asociados con predisposición a LES
• Keir ME, Francisco LM, Sharpe AH. PD-1 and its ligands in T-cell immunity.
Curr Opin Immunol. 2007; 19: 309-314.
• Kel AE, Gossling E, Reuter I, Cheremushkin E, Kel-Margoulis OV,
Wingender E et al. MATCH: A tool for searching transcription factor binding
sites in DNA sequences. Nucleic Acids Res. 2003; 31: 3576-3579.
• Kelly JA, Kelley JM, Kaufman KM, Kilpatrick J, Bruner GR, Merrill JT et al.
Interferon regulatory factor-5 is genetically associated with systemic lupus
erythematosus in African Americans. Genes Immun. 2008; 9: 187-194.
• Kelly JA, Moser KL, Harley JB. The genetics of systemic lupus erythematosus:
putting the pieces together. Genes Immun. 2002; 3 Suppl 1:S71-S85.
• Kim-Howard X, Maiti AK, Anaya JM, Bruner GR, Brown E, Merrill JT et al.
ITGAM coding variant (rs1143679) influences the risk of renal disease, discoid
rash, and immunologic manifestations in lupus patients with European ancestry.
Ann Rheum Dis. 2009. [Epub ahead of print].
• Klinman DM, Gursel I, Klaschik S, Dong L, Currie D, Shirota H. Therapeutic
potential of oligonucleotides expressing immunosuppressive TTAGGG motifs.
Ann N Y Acad Sci. 2005; 1058: 87-95.
• Klose RJ, Bird AP. Genomic DNA methylation: the mark and its mediators.
Trends Biochem Sci. 2006; 31: 89-97.
• Kobayashi S, Ikari K, Kaneko H, Kochi Y, Yamamoto K, Shimane K et al.
Association of STAT4 with susceptibility to rheumatoid arthritis and systemic
lupus erythematosus in the Japanese population. Arthritis Rheum. 2008; 58:
1940-1946.
• Korman BD, Alba MI, Le JM, Alevizos I, Smith JA, Nikolov NP et al.
Variant form of STAT4 is associated with primary Sjogren's syndrome. Genes
Immun. 2008; 9:267-270.
256
Bibliografía
257
Factores genéticos asociados con predisposición a LES
• Lee YH, Nath SK. Systemic lupus erythematosus susceptibility loci defined by
genome scan meta-analysis. Hum Genet. 2005; 118: 434-443.
• Lee-Kirsch MA, Gong M, Chowdhury D, Senenko L, Engel K, Lee YA et al.
Mutations in the gene encoding the 3'-5' DNA exonuclease TREX1 are
associated with systemic lupus erythematosus. Nat Genet. 2007; 39: 1065-1067.
• Leibson PJ. The regulation of lymphocyte activation by inhibitory receptors.
Curr Opin Immunol. 2004; 16: 328-336.
• Li CC, Ramirez-Carrozzi VR, Smale ST. Pursuing gene regulation 'logic' via
RNA interference and chromatin immunoprecipitation. Nat Immunol. 2006; 7:
692-697.
• Li Q, Johnston SA. Are all DNA binding and transcription regulation by an
activator physiologically relevant? Mol Cell Biol. 2001; 21: 2467-2474.
• Li QJ, Chau J, Ebert PJ, Sylvester G, Min H, Liu G et al. miR-181a is an
intrinsic modulator of T cell sensitivity and selection. Cell. 2007; 129: 147-161.
• Lindqvist AK, Steinsson K, Johanneson B, Kristjansdottir H, Arnasson A,
Grondal G et al. A susceptibility locus for human systemic lupus erythematosus
(hSLE1) on chromosome 2q. J Autoimmun. 2000; 14: 169-178.
• MacKenzie A, Quinn J. A serotonin transporter gene intron 2 polymorphic
region, correlated with affective disorders, has allele-dependent differential
enhancer-like properties in the mouse embryo. Proc Natl Acad Sci U S A. 1999;
96: 15251-15255.
• Magnusson V, Lindqvist AK, Castillejo-Lopez C, Kristjansdottir H, Steinsson
K, Grondal G et al. Fine mapping of the SLEB2 locus involved in susceptibility
to systemic lupus erythematosus. Genomics. 2000; 70: 307-314.
• Manderson AP, Botto M, Walport MJ. The role of complement in the
development of systemic lupus erythematosus. Annu Rev Immunol. 2004; 22:
431-456.
258
Bibliografía
• Manolio TA, Collins FS, Cox NJ, Goldstein DB, Hindorff LA, Hunter DJ et
al. Finding the missing heritability of complex diseases. Nature. 2009; 461:
747-753.
• Martinez A, Varade J, Marquez A, Cenit MC, Espino L, Perdigones N et al.
Association of the STAT4 gene with increased susceptibility for some immune-
mediated diseases. Arthritis Rheum. 2008; 58: 2598-2602.
• Mevorach D, Mascarenhas JO, Gershov D, Elkon KB. Complement-
dependent clearance of apoptotic cells by human macrophages. J Exp Med.
1998; 188: 2313-2320.
• Meyers S, Downing JR, Hiebert SW. Identification of AML-1 and the (8;21)
translocation protein (AML-1/ETO) as sequence-specific DNA-binding proteins:
the runt homology domain is required for DNA binding and protein-protein
interactions. Mol Cell Biol. 1993; 13: 6336-6345.
• Miller W, Rosenbloom K, Hardison RC, Hou M, Taylor J, Raney B et al. 28-
way vertebrate alignment and conservation track in the UCSC Genome
Browser. Genome Res. 2007; 17: 1797-1808.
• Minegishi Y, Saito M, Morio T, Watanabe K, Agematsu K, Tsuchiya S et al.
Human tyrosine kinase 2 deficiency reveals its requisite roles in multiple
cytokine signals involved in innate and acquired immunity. Immunity 2006; 25:
745-755.
• Mishra N, Reilly CM, Brown DR, Ruiz P, Gilkeson GS. Histone deacetylase
inhibitors modulate renal disease in the MRL- lpr/ lpr mouse. J Clin Invest.
2003; 111: 539-552.
• Mok CC, Lau CS. Pathogenesis of systemic lupus erythematosus. J Clin
Pathol. 2003; 56: 481-490.
• Moser KL, Kelly JA, Lessard CJ Harley JB. Recent insights into the genetic
basis of systemic lupus erythematosus. Genes Immun. 2009; 10: 373-379.
259
Factores genéticos asociados con predisposición a LES
260
Bibliografía
• Nepom GT, Byers P, Seyfried C, Healey LA, Wilske KR, Stage D et al. HLA
genes associated with rheumatoid arthritis: identification of susceptibility
alleles using specific oligonucleotide probes. Arthritis Rheum. 1989; 32: 15-21.
• Newby PR, Roberts-Davies EL, Brand OJ, Heward JM, Franklyn JA, Gough
SC et al. Tag SNP screening of the PDCD1 gene for association with Graves'
disease. Clin Endocrinol (Oxf). 2007; 67: 125-128.
• Nielsen C, Hansen D, Husby S, Jacobsen BB, Lillevang ST. Association of a
putative regulatory polymorphism in the PD-1 gene with susceptibility to type 1
diabetes. Tissue Antigens. 2003; 62: 492-497.
• Nielsen C, Laustrup H, Voss A, Junker P, Husby S, Lillevang ST. A putative
regulatory polymorphism in PD-1 is associated with nephropathy in a
population-based cohort of systemic lupus erythematosus patients. Lupus. 2004;
13: 510-516.
• Niewold TB, Kelly JA, Flesch MH, Espinoza LR, Harley JB and Crow MK.
Association of the IRF5 risk haplotype with high serum interferon-alpha activity
in systemic lupus erythematosus patients. Arthritis Rheum. 2008; 58 (8): 2481-
2487.
• Nishikomori R, Usui T, Wu CY, Morinobu A, O'Shea JJ, Strober W.
Activated STAT4 has an essential role in Th1 differentiation and proliferation
that is independent of its role in the maintenance of IL-12R beta 2 chain
expression and signaling. J Immunol. 2002; 169: 4388-4398.
• Nishimura H, Minato N, Nakano T, Honjo T. Immunological studies on PD-1
deficient mice: implication of PD-1 as a negative regulator for B cell responses.
Int Immunol. 1998; 10: 1563-1572.
• Nishimura H, Nose M, Hiai H, Minato N, Honjo T. Development of lupus-like
autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-
carrying immunoreceptor. Immunity. 1999; 11: 141-151.
261
Factores genéticos asociados con predisposición a LES
262
Bibliografía
263
Factores genéticos asociados con predisposición a LES
• Petrovas C, Casazza JP, Brenchley JM, Price DA, Gostick E, Adams WC et al.
PD-1 is a regulator of virus-specific CD8+ T cell survival in HIV infection. J
Exp Med. 2006; 203 (10): 2281-2292
• Plenge RM, Cotsapas C, Davies L, Price AL, de Bakker PIW, Maller J et al.
Two independent alleles at 6q23 associated with risk of rheumatoid arthritis.
Nat Genet. 2007a; 39: 1477-1482.
• Plenge RM, Padyukov L, Remmers EF, Purcell S, Lee AT, Karlson EW et al.
Replication of putative candidate-gene associations with rheumatoid arthritis in
4,000 samples from North America and Sweden: association of susceptibility
with PTPN22, CTLA4, and PADI4. Am J Hum Genet. 2005; 77: 1044-1060.
• Plenge RM, Seielstad M, Padyukov L, Lee AT, Remmers EF, Ding B et al.
TRAF1-C5 as a risk locus for rheumatoid arthritis: a genomewide study. N
Engl J Med. 2007b; 357: 1199-1209.
• Pons-Estel BA, Catoggio LJ, Cardiel MH, Soriano ER, Gentiletti S, Villa AR
et al. The GLADEL multinational Latin American prospective inception cohort
of 1,214 patients with systemic lupus erythematosus: ethnic and disease
heterogeneity among "Hispanics". Medicine (Baltimore). 2004; 83: 1-17.
• Price AL, Patterson NJ, Plenge RM, Weinblatt ME, Shadick NA, Reich D.
Principal components analysis corrects for stratification in genome-wide
association studies. Nat Genet. 2006; 38: 904-909.
• Prokunina L, Castillejo-Lopez C, Oberg F, Gunnarsson I, Berg L, Magnusson
V et al. A regulatory polymorphism in PDCD1 is associated with susceptibility
to systemic lupus erythematosus in humans. Nat Genet. 2002; 32: 666-669.
• Prokunina L, Gunnarsson I, Sturfelt G, Truedsson L, Seligman VA, Olson JL
et al. The systemic lupus erythematosus-associated PDCD1 polymorphism
PD1.3A in lupus nephritis. Arthritis Rheum. 2004; 50: 327-328.
• Radziewicz H, Dunham RM, Grakoui A. PD-1 tempers Tregs in chronic HCV
infection. J Clin Invest. 2009; 119: 450-453.
264
Bibliografía
265
Factores genéticos asociados con predisposición a LES
• Ren Y, Tang J, Mok MY, Chan AW, Wu A, Lau CS. Increased apoptotic
neutrophils and macrophages and impaired macrophage phagocytic clearance
of apoptotic neutrophils in systemic lupus erythematosus. Arthritis Rheum.
2003; 48: 2888-2897.
• Reveille JD, Arnett FC, Wilson RW, Bias WB, McLean RH. Null alleles of
the fourth component of complement and HLA haplotypes in familial systemic
lupus erythematosus. Immunogenetics. 1985; 21: 299-311.
• Richardson B, Scheinbart L, Strahler J, Gross L, Hanash S, Johnson M.
Evidence for impaired T cell DNA methylation in systemic lupus erythematosus
and rheumatoid arthritis. Arthritis Rheum. 1990; 33: 1665-1673.
• Richman IB, Chung SA, Taylor KE, Kosoy R, Tian C, Ortmann WA et al.
European population substructure correlates with systemic lupus erythematosus
endophenotypes in North Americans of European descent. Genes Immun. 2009.
[Epub ahead of print].
• Richter MF, Dumenil G, Uze G, Fellous M, Pellegrini S. Specific contribution
of TYK2 JH regions to the binding and the expression of the interferon a/b
receptor component IFNAR1. J Biol Chem. 1998; 273: 24723-24729.
• Rieck M, Arechiga A, Onengut-Gumuscu S, Greenbaum C, Concannon P,
Buckner JH. Genetic variation in PTPN22 corresponds to altered function of T
and B lymphocytes. J Immunol. 2007; 179: 4704-4710.
• Roulet E, Fisch I, Junier T, Bucher P, Mermod N. Evaluation of computer
tools for the prediction of transcription factor binding sites on genomic DNA. In
Silico Biol. 1998; 1: 21-28.
• Salloum R, Franek BS, Kariuki SN, Rhee L, Mikolaitis RA, Jolly M et al.
Genetic variation at the IRF7/PHRF1 locus is associated with autoantibody
profile and serum interferon-alpha activity in lupus patients. Arthritis Rheum.
2010; 62: 553-561.
266
Bibliografía
267
Factores genéticos asociados con predisposición a LES
• Shappell SB, Toman C, Anderson DC, Taylor AA, Entman ML, Smith CW.
Mac-1 (CD11b/CD18) mediates adherence-dependent hydrogen peroxide
production by human and canine neutrophils. J Immunol. 1990; 144: 2702-
2711.
• Sharpe AH, Freeman GJ. The B7-CD28 superfamily. Nat Rev Immunol. 2002;
2: 116-126.
• Shimane K, Kochi Y, Horita T, Ikari K, Amano H, Hirakata M et al. The
association of a nonsynonymous single-nucleotide polymorphism in TNFAIP3
with systemic lupus erythematosus and rheumatoid arthritis in the Japanese
population. Arthritis Rheum. 2010; 62: 574-579.
• Shin HD, Kim I, Choi CB, Lee SO, Lee HW, Bae SC. Different genetic
effects of interferon regulatory factor 5 (IRF5) polymorphisms on systemic
lupus erythematosus in a Korean population. J Rheumatol. 2008; 35: 2148-
2151.
• Sigurdsson S, Göring HH, Kristjansdottir G, Milani L, Nordmark G, Sandling
JK et al. Comprehensive evaluation of the genetic variants of interferon
regulatory factor 5 (IRF5) reveals a novel 5 bp length polymorphism as strong
risk factor for systemic lupus erythematosus. Hum Mol Genet. 2008a; 17: 872-
881.
• Sigurdsson S, Nordmark G, Garnier S, Grundberg E, Kwan T, Nilsson O et al.
A risk haplotype of STAT4 for systemic lupus erythematosus is over-expressed,
correlates with antidsDNA and shows additive effects with two risk alleles of
IRF5. Hum Mol Genet. 2008b; 17: 2868-2876.
• Sigurdsson S, Nordmark G, Göring HH, Lindroos K, Wiman AC, Sturfelt G,
et al. Polymorphisms in the Tyrosine Kinase 2 and Interferon Regulatory
Factor 5 Genes Are Associated with Systemic Lupus Erythematosus. Am J Hum
Genet. 2005. 76: 528-537.
268
Bibliografía
269
Factores genéticos asociados con predisposición a LES
270
Bibliografía
271
Factores genéticos asociados con predisposición a LES
272
Bibliografía
273
Factores genéticos asociados con predisposición a LES
CBF binding sites in promoter activation. Proc Natl Acad Sci U S A. 1995; 92:
6930-6934.
• Wasserman R, Li YS, Hardy RR. Differential expression of the blk and ret
tyrosine kinases during B lineage development is dependent on Ig
rearrangement. J Immunol. 1995; 155: 644-651.
• Watford WT, Hissong BD, Bream JH, Kanno Y, Muul L, O’Shea JJ.
Signaling by IL-12 and IL-23 and the immunoregulatory roles of STAT4.
Immunol Rev. 2004; 202: 139-156.
• Webb R, Wren JD, Jeffries M, Kelly JA, Kaufman KM, Tang Y et al.
Variants within MECP2, a key transcription regulator, are associated with
increased susceptibility to lupus and differential gene expression in patients
with systemic lupus erythematosus. Arthritis Rheum. 2009; 60: 1076-1084.
• Wellcome Trust Case Control Consortium. Genome-wide association study of
14,000 cases of seven common diseases and 3,000 shared controls. Nature.
2007; 447: 661-678.
• Wen ZK, Xu W, Xu L, Cao QH, Wang Y, Chu YW et al. DNA
hypomethylation is crucial for apoptotic DNA to induce systemic lupus
erythematosus-like autoimmune disease in SLE-non-susceptible mice.
Rheumatology (Oxford). 2007; 46: 1796-1803.
• Wertz IE, O'Rourke KM, Zhou H, Eby M, Aravind L, Seshagiri S et al. De-
ubiquitination and ubiquitin ligase domains of A20 downregulate NF-kappaB
signalling. Nature. 2004; 430: 694-699.
• Willcocks LC, Lyons PA, Clatworthy MR, Robinson JI, Yang W, Newland
SA et al. Copy number of FCGR3B, which is associated with systemic lupus
erythematosus, correlates with protein expression and immune complex uptake.
J Exp Med. 2008; 205: 1573-1582.
• Wilson WA, Gharavi AE, Koike T, Lockshin MD, Branch DW, Piette JC et al.
International consensus statement on preliminary classification criteria for
274
Bibliografía
275
Factores genéticos asociados con predisposición a LES
276
ANEXO
Anexo
1
Laboratorio Investigacion 10 and Rheumatology Unit, Hospital Clinico Universitario
de Santiago, Santiago de Compostela. Spain; [email protected],
[email protected], [email protected]
2
Department of Medicine. University of Santiago de Compostela, Santiago de
Compostela. Spain; [email protected]
* These authors contributed equally
Corresponding author:
Antonio Gonzalez
Laboratorio de Investigacion 10
15706-Santiago de Compostela
Spain
279
Factores genéticos asociados con predisposición a LES
280
Anexo
ABSTRACT
Introduction: We aimed to replicate the strong associations that a recent Genome Wide
Association Study (GWAS) has found between 16 SNPs and response to anti-TNF
treatment in 89 patients with RA. This study is very important because, according with
published simulations, associations as strong as the reported will mean that these SNPs
Methods: Disease activity score (DAS28) was evaluated in 151 anti-TNF treated
Association between the relative change in DAS28 and SNP genotypes was tested by
response criteria.
Results: None of the analyses showed any significant association between the 16 SNPs
and response to anti-TNF treatments at 3 or 6 months. Results were also negative when
only patients treated with Infliximab (66.9 % of the total) were separately analyzed.
These negative results were obtained in spite of a very good statistical power to
Conclusion: We still do not have any sound evidence of genetic variants associated
of using the results of a recent GWAS for prediction in individual patients should be
dismissed.
281
Factores genéticos asociados con predisposición a LES
INTRODUCTION
rheumatoid arthritis (RA)[1-2]. Three drugs of this type, Infliximab, Etanercept and
Adalimumab, have been used with success in hundreds of thousands patients with RA
around the world. New drugs targeting TNF are in development or have been recently
approved[3]. The beneficial effects of these drugs include a better quality of life, control
of inflammation, stiffness and pain, and slowing progression to joint erosions and
deformity. It seems also that they are able to decrease cardiovascular risk and overall
patients that do not obtain these advantageous effects[1-3]. In some of them, this lack of
response is primary, from the start of the treatment, whereas others develop resistance to
treatment after a period of initial response. Unfortunately, there are no useful predictors
to forecast how the clinical response of a specific patient will be. This has led to an
unsatisfactory trial and error approach in the selection of drugs, which means that some
patients will miss an effective treatment at a critical window of opportunity time[6] and
a waste of resources for the health services. As a response to this challenge, multiple
lines of research are looking for predictors of response to anti-TNF therapies among
this last field. Sixteen SNPs with an important association with response to treatment
were identified in a recent Genome Wide Association (GWA) study[7]. In our view, the
most remarkable aspect of these findings was the marked effect size of each SNP, with
levels very rarely found in genetic studies of complex traits. All showed O.R. over 3.5
in the comparison between patients with good response and non-responders. Some of
them showed effect sizes over O.R. = 10. These effects together with minor allele
282
Anexo
frequencies over 12 % mean that they will allow, if confirmed, for prediction of
response to anti-TNF treatments with great accuracy at the level of the individual
patient[11]. The limitation of this study was that only 89 patients were included and
even very significant results in a study of this size are uncertain. Our objective has been
to provide the necessary replication to these exciting findings with the expectation that,
at least, a few of them will be confirmed. This will be a first step before proceeding to
efficacy of anti-TNF therapy. All of them were of European (Spanish) ancestry. Only
patients that were naïve with respect to biologic treatments were included. Patients have
been systematically evaluated at the initiation of therapy and every three months
thereafter. Evaluations included painful and swollen joint counts, visual analogue scales
of pain, global health assessment by the patient and the physician, erythrocyte
(HAQ) and DAS-28 score. Clinical characteristics are detailed in Table 1. All
participants gave their informed consent for inclusion and the study and procedures
have been approved by the Ethics Committee for Clinical Research of Galicia.
Assessment of the efficacy of the treatment: We used the same procedures described
in Liu et al. to make our results comparable[7]. Response to anti-TNF treatments was
assessed with the DAS28 score.[12] The primary outcome was the quantitative variable
283
Factores genéticos asociados con predisposición a LES
3.2; moderate-responders have ∆DAS28 ≥ 1.2 and DAS28 at 3 months > 3.2 or 0.6 <
∆DAS28 ≤ 1.2 and DAS28 at 3 months ≤ 5.1; and non-responders are those who do not
Genotypes: A total of 16 SNPs from Liu et al. [7] were analyzed (Table 2). Genotypes
were obtained by single-base extension with the SNaPshot Multiplex Kit (Applied
Biosystems, Foster City, CA, USA) and specific primers and probes (available as
supplementary material). Genotype call rate was 99.79 %, allele frequencies were in
Hardy-Weinberg equilibrium and concordant results for the 16 SNPs were obtained in
included in the GWAS and in our study was done with the Student t test for data
available as means and standard deviation and with the Chi squared test for contingency
tables for frequency data. Analyses of the relationship between SNPs and treatment
response were done as in Liu et al. to make our results comparable also in this aspect[7].
Briefly, linear regression analysis between genotype data following a genetic additive
model and relDAS28 as the continuous dependent variable was done. A t statistic was
derived from the linear regression and used to calculate the p-value of the association.
logistic regression analysis between the groups of responders and non-responders. Odds
ratios (O.R.) and their 95 % confidence intervals (C.I.) were obtained using as reference
the non-responder group. This second analysis will be less powerful because the
284
Anexo
a customized version of the Statistica 7.0 program (StatSoft, Tulsa, OK, USA). We also
explored visually the possibility that consideration of all the SNPs jointly will
discriminate between responder and non-responder patients. This analysis was done
with the Co-Plot algorithm implemented in the Visual Co-Plot software (D. Talby and
Estimation of the statistical power for the linear regression analysis was done by
correlation coefficients (R2). The values of R2 and the number of samples in our study
were used as input in the module for the F test in omnibus comparisons by linear
RESULTS
The aim of our study was to replicate the strong association of 16 SNPs with
the same variables and type of analysis. Data from the 151 patients with RA are shown
in table 1. Some of the characteristics of our study population were different from the
patients analyzed in the GWAS[7]. Specifically, our patients showed a lower percentage
of RF positivity, higher positivity for anti-CCP antibodies, and higher baseline HAQ
and DAS28 levels. There were 70.2 % patients with high disease activity at baseline as
assessed by a DAS28 > 5.1. In spite of this high activity, there were 40.4 % and 43.7 %
good responders at 3 and 6 months, respectively, and only 22.5 % and 21.8 % of non-
responders were similar in the two studies. In contrast, the proportion of patients treated
with each of the three anti-TNF drugs was different (Table 1). In our cohort, most
patients were treated with Infliximab (66.9 %), followed by Etanercept (23.2 %) and
285
Factores genéticos asociados con predisposición a LES
Adalimumab (9.9 %). We also checked that there was a good correlation between the
variable used as primary outcome in our analysis, redDAS28, and the EULAR response
The relationship between the SNP genotypes and response to anti-TNF treatment
at 3 months was evaluated by linear regression analysis between the genotypes and the
continuous variable relDAS28, which is the relative change in DAS28 between baseline
and the time of evaluation. There was not association of any of the 16 SNPs with
relDAS28 at 3 months (Table 2). Secondary analyses showed very similar results.
at 3 months did not show any significant association (Table 2). The most extreme O.R.
= 1.9 (95 % C.I. = 1.0-3.6) corresponded to rs437943 in the CNTDE1 locus, but it
compared poorly with the previously reported O.R. = 4.6 (95 % C.I. = 1.8-12.3). In
addition, there was not association of relDAS28 with any of the 16 SNPs at 6 months,
represented 66.9 % of the patients in our study, did not show any significant association
Etarnercept or Adalimumab were done because of the small number of patients in these
two subgroups. We also visually explored if joint consideration of the 16 SNPs was able
treatment, but patients with different responses did not show any clustering in
It was critical, for interpretation of the above results, to assess if our study had
enough statistical power to replicate the previously reported associations. Power for the
286
Anexo
larger than 95 % for a P value of 0.002. It is important to remark that O.R. from the
GWAS are very likely heavily biased upwards as consequence of the winner’s curse
affecting any GWAS and specially those of small size [16-17]. Therefore this power
estimate is only valuable in the context of the reported O.R. taken at face value.
DISCUSSION
treatments[1-3, 9]. The development and approval of new effective drugs for RA adds
to this urgency[3]. The recent GWAS from Liu et al. [7] was especially remarkable
they could be used for prediction in individual patients. This is a characteristic that has
not been found in any of the previous studies. However, the size of the study implied
that results should be replicated before they could be taken at face value, as already
acknowledged by the authors. We have tried to provide here the needed replication in
the expectation that some of them will be confirmed and that validation in prospective
larger sample size of our study and the corresponding very good power to detect this
type of strong associations. These results make very unlikely that any of the 16 SNPs
possible that the differences between the patients with RA in the two studies could have
had an effect in the lack of replication, but these differences were not large enough to
completely explain the very divergent results. In addition, patients in the GWAS were
predominantly of European ancestry as were all the patients in our study. Therefore, it
seems more likely that the original strong associations were due to random variation of
287
Factores genéticos asociados con predisposición a LES
allele frequencies in a study including more than 300 000 SNPs and to the heavy bias
characteristic of GWAS of small sample size [16-17]. This possibility was already
considered by us before beginning this study, but we judged that some SNPs will be
replicated given that they showed low P values, 5 of them with P values below 10-6[7],
and low P values are the best indication of the reproducibility of results[18].
CONCLUSIONS
Our negative results implicate that we do not have still any strong evidence
addition, our results imply that none of the SNPs in our study will be useful as
that they will show association with response to anti-TNF, but of a much weaker
strength.
ABBREVATIONS
10
288
Anexo
COMPETING INTERESTS
Roche Spain contributed to the funding of this project. However, the company
had no input in the design of the study or in the analysis and writing of the manuscript.
The company did not have the right to early access to results or to interfere in any other
way with the interpretation or reporting of the results. Therefore, the authors keep
AUTHORS’ CONTRIBUTION
the interpretation of the results and in writing the manuscript. EP-P participated in the
acquisition of clinical data and collection of samples and in the analysis and
the study and in the coordination of acquisition of clinical data and collection of
of the manuscript. All authors read and approved the final manuscript
ACKNOWLEDGMENTS
We thank Carmen Pena-Pena for her excellent technical assistance and Yolanda
Lopez-Golan for her help in recruiting patients. M.S-G is the recipient of a FPU pre-
“Isabel Barreto” bursary of the Government of Galicia. This project was supported by
an unrestricted grant from Roche Spain and by grants PI080744 and PI09/90744 form
11
289
Factores genéticos asociados con predisposición a LES
the Instituto de Salud Carlos III (Spain) with participation of funds from FEDER
(European Union).
REFERENCES
1. Chen YF, Jobanputra P, Barton P, Jowett S, Bryan S, Clark W, Fry-Smith A,
arthritis failing initial TNF inhibitor therapy: a critical review. Arthritis Res
arthritis are not greater than expected when treated with tumour necrosis
5. Dixon WG, Watson KD, Lunt M, Hyrich KL, Silman AJ, Symmons DP:
12
290
Anexo
A, Kern M, Furie R, Dupuis J, Plenge RM, Coenen MJ, Behrens TW, Carulli JP,
8. Maxwell JR, Potter C, Hyrich KL, Barton A, Worthington J, Isaacs JD, Morgan
AW, Wilson AG: Association of the tumour necrosis factor-308 variant with
arthritis in the era of biologic agents. Nat Rev Rheumatol 2009, 5(9):491-496.
10. Plenge RM, Criswell LA: Genetic variants that predict response to anti-
11. Janssens AC, Moonesinghe R, Yang Q, Steyerberg EW, van Duijn CM, Khoury
profiling for predicting common chronic diseases. Genet Med 2007, 9(8):528-
535.
12. Prevoo ML, van 't Hof MA, Kuper HH, van Leeuwen MA, van de Putte LB, van
13. van Gestel AM, Prevoo ML, van 't Hof MA, van Rijswijk MH, van de Putte LB,
van Riel PL: Development and validation of the European League Against
13
291
Factores genéticos asociados con predisposición a LES
14. Bravata DM, Shojania KG, Olkin I, Raveh A: CoPlot: a tool for visualizing
power analysis program for the social, behavioral, and biomedical sciences.
16. Ioannidis JP: Why most discovered true associations are inflated.
17. Xiao R, Boehnke M: Quantifying and correcting for the winner's curse in
18. Todd JA: Statistical false positive or true disease pathway? Nat Genet 2006,
38(7):731-733.
14
292
Anexo
FIGURE LEGENDS
classification and relDAS28. Median, interquartile range, and non-outlier range are
represented as dots, box and whiskers, respectively. Empty dots represent outliers.
Figure 2: Multivariate visual analysis showing that 16 SNPs were not able to
represented as blue dots; moderate responders as red dots; and non-responders as green
dots. Yellow arrows represent the genotypes of each of the 16 SNPs according to an
15
293
Factores genéticos asociados con predisposición a LES
Table 1. Clinical characteristics of the patients in this study compared with the report
from Liu et al. [7]
Variable This study Data from ref. 7 P-value
Age (mean ± SD) 54.2 ± 13.1 57 ± 13.5 0.06
Age at diagnosis (mean ± SD) 46.2 ± 13.5 47 ± 15 0.3
Women (%) 84.8 75 0.06
Disease duration (mean ± SD) 7.8 ± 6.6 8±8 0.4
a
Current Smokers (%) 9.1 15 0.3
a
Ever Smokers (%) 25.0
anti-TNF drug
Infliximab (n) 101 32 0.01
Etanercept (n) 35 39 0.02
Adalimumab (n) 15 18 0.05
a
Data was available for 102 patients for CCP antibodies, ANA and SE; and for 88 patients for smoking
habits.
16
294
Anexo
Table 2. Relationship of relDAS28 and SNP genotypes, and comparison of the allele
frequencies between responders and non-responders.
Responders’
P value of Non-responders’ MAF % c P
SNP MAF % b O.R. (95% C.I.)
relDAS28 b (n/N) value
(n/N)
rs983332 0.6 25.9 (30/116) 27.9 (19/68) 1,11 (0,6-2,0) 0.8
rs928655 0.1 18.9 (23/122) 28.0 (19/68) 0.60 (0.3-1.2) 0.1
rs13393173 0.8 23.8 (29/122) 20.6 (14/68) 0,83 (0,4-1,7) 0.6
rs437943 0.05 38.5 (47/122) 25.0 (17/68) 0,53 (0,3-1,0) 0.06
rs10945919 0.7 27.9 (34/122) 23.5 (16/68) 0,79 (0,4-1,7) 0.5
rs854547 0.3 39.3 (48/122) 36.8 (25/68) 1.12 (0.6-2.1) 0.7
rs854548 0.9 23.0 (28/122) 23.5 (16/68) 1,03 (0,5-2,0) 0.9
rs854555 0.6 35.2 (43/122) 38.2 (26/68) 1,14 (0,6-2,0) 0.7
rs868856 0.6 32.8 (40/122) 36.8 (25/68) 1,19 (0,6-2,0) 0.6
rs7046653 0.5 32.0 (39/122) 36.8 (25/68) 1,23 (0,7-2,5) 0.5
rs2814707 0.8 25.4 (31/122) 29.4 (20/68) 1,22 (0,6-2,5) 0.6
rs3849942 0.4 23.0 (28/122) 29.4 (20/68) 1,41 (0,7-2,5) 0.3
rs774359 0.2 26.2 (32/122) 36.8 (25/68) 1,64 (0,8-3,3) 0.1
rs6138150 0.5 14.0 (17/122) 14.7 (10/68) 0.94 (0.4-2.2) 0.9
rs6028945 0.9 12.3 (15/122) 13.2 (9/68) 1,09 (0,5-2,5) 0.9
rs6071980 0.9 18.9 (23/122) 16.2 (11/68) 0,83 (0,4-2,0) 0.6
a
Major/Minor alleles
b
MAF = Minor Allele Frequency, n = number of minor alleles, N = total number of alleles
c
O.R. were calculated as in Liu et al [7], taking the allele associated with the non-responders as the
numerator of the non-responders odds, and the non-responders odds as the numerator of the O.R.
17
295
Factores genéticos asociados con predisposición a LES
Figure 1
296
Anexo
Figure 2
297
298
Supplementary Table 1. Primers and probes in 5’ĺ 3’ direction used for genotyping the 16 SNPs by the single-base extension approach.
PCR primers
SNP Forward Primer Reverse Primer Minisequencing Probe
rs983332 atctgtagctctgtttctgcttgg caagtactccttgctgccaaaag cagactctctataattttaatggaatcaatacacaaggtccagcgtccaacta
rs928655 gcctttcccatcacattctc ttctcagctgcctccttctt CCAGAAGTATATTAATGAGCAGTGCgtgaattctgtgatttctcaccctgcctttcagatttg $
rs13393173 tggtgttctcattgccttgaatg gaacaagatggtctctatcccagg tggaaagccctCagacagctacaatttagctaactcca
rs437943 ccagcattggtggaagtgat tggtcagtttgaaaggtctgc TTACCTATGATTGATgagcaccatatagaacttatagcaacca $
rs10945919 tcactgcagttctgcctttg tgatccactttgtgtttgga tttggaaatttactattgaaagagaaaggttcaaagctgaaacattagctgcccaaggactca
rs854547 aagtggtcctcctgcttcag ttggaccaagactagatttcctg gctgcttttcaggagtgaatcactgaag
rs854548 aacaagccttgactgggtaaac tgcaggctttgctcttttaac TTACCTATGAtgactgggtaaacctgtatcaattgtcaaagGatatatctgtcaatca $
rs854555 cccaagagatacgggctttaac tttgtccacggtctttattcag catcatatatatgatatcatcttgacatatttgtctactgctg
rs868856 cgctggtgcttcatactcac agcctttgcagagacacctg TTACCTATGAtcctagtccgtctttctctttcacttgtggttttcttcccttgtcatttaagcattctcaact $
rs7046653 tgtaggcacaatgggtagctt gagacactaggaccgtttgttg tcctggtatcctttattttctgaattaaaaaaaaaaaaggctgtatga
rs2814707 aaacgacaagagaagcacagc gcccacctcaaactaaagca aattcctgcatccagtagtgcccaaagccagtg
rs3849942 ccatgctaggcactgagacac cccactacagcccattcttc ctttcttcccacaggtctagctagtacgtatttctttt
rs774359 agtaaaggaactaacatgtaggcactc gtacccgtgggcaaggtg cacctatttcctcatctgtaaaatggcaataatagtaatagtacctaatgtgt
Factores genéticos asociados con predisposición a LES
Primers selected with Primer3 and Oligos softwares. They were checked to avoid formation of dimers between the primers included in the reaction.
$ These oligonucleotides were extended with a 5' tail that has not homology with human sequences (these tails are in capital letters)
Anexo
a
MAF = Minor Allele Frequency, n = number of minor alleles, N = total number of alleles
299
Factores genéticos asociados con predisposición a LES
a
MAF = Minor Allele Frequency, n = number of minor alleles, N = total number of alleles
300