87 results on '"Kerick, M"'
Search Results
2. GWAS for systemic sclerosis identifies multiple risk loci and highlights fibrotic and vasculopathy pathways
- Author
-
López-Isac, E., Acosta-Herrera, M., Kerick, M., Assassi, S., Satpathy, A.T., Granja, J., Mumbach, M.R., Beretta, L., Simeón, C.P., Carreira, P., Ortego-Centeno, N., Castellvi, I., Bossini-Castillo, L., Carmona, F.D., Orozco, G., Hunzelmann, N., Distler, J.H.W., Franke, A., Lunardi, C., Moroncini, G., Gabrielli, A., de Vries-Bouwstra, J., Wijmenga, C., Koeleman, B.P.C., Nordin, A., Padyukov, L., Hoffmann-Vold, A.-M., Lie, B., Ríos, R., Callejas, J.L., Vargas-Hitos, J.A., García-Portales, R., Camps, M.T., Fernández-Nebro, A., González-Escribano, M.F., García-Hernández, F.J., Castillo, M.J., Aguirre, M.A., Gómez-Gracia, I., Fernández-Gutiérrez, B., Rodríguez-Rodríguez, L., García de la Peña, P., Vicente, E., Andreu, J.L., Fernández de Castro, M., López-Longo, F.J., Martínez, L., Fonollosa, V., Guillén, A., Espinosa, G., Tolosa, C., Pros, A., Rodríguez-Carballeira, M., Narváez, F.J., Rubio-Rivas, M., Ortiz-Santamaría, V, Madroñero, A.B., González-Gay, M.A., Díaz, B., Trapiella, L., Sousa, A., Egurbide, M.V., Fanlo-Mateo, P., Sáez-Comet, L., Díaz, F., Hernández, V, Beltrán, E., Román-Ivorra, J.A., Grau E., Alegre-Sancho, J.J., Freire, M., Blanco-García, F.J., Oreiro, N., Witte, T., Kreuter, A., Riemekasten, G., Airó P., Magro, C., Voskuyl, A.E., Vonk, M.C., Hesselstrand, R., Proudman S., Stevens, W., Nikpour, M., Zochling, J., Sahhar, J., Roddy, J., Nash, P., Tymms, K., Rischmueller, M., Lester, S., Vyse, T., Herrick, A.L., Worthington, J., Denton C.P., Allanore, Y., Brown, M.A., Radstake, T.R.D.J., Fonseca, C., Chang H.Y., Mayes, M.D., Martin, J., European Scleroderma Group, Australian Scleroderma Interest Group (ASIG), Universidad de Salamanca, Ministerio de Economía y Competitividad (España), Junta de Andalucía, Ministerio de Educación, Cultura y Deporte (España), Instituto de Salud Carlos III, Howard Hughes Medical Institute, Federal Ministry of Education and Research (Germany), Universidad de Cantabria, [López-Isac E, Acosta-Herrera M, Kerick M] Institute of Parasitology and Biomedicine López-Neyra, IPBLN-CSIC, Granada, Spain. [Assassi S] The University of Texas Health Science Center-Houston, Houston, USA. [Satpathy AT, Granja J] Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, USA. Howard Hughes Medical Institute, Stanford University, Stanford, USA. [Simeón CP] Servei de Medicina Interna, Vall d'Hebron Hospital Universitari, Barcelona, Spain, Vall d'Hebron Barcelona Hospital Campus, Groningen Institute for Gastro Intestinal Genetics and Immunology (3GI), Rheumatology, and AII - Inflammatory diseases
- Subjects
0301 basic medicine ,Chemistry(all) ,AUTOIMMUNITY ,Àcids nucleics ,General Physics and Astronomy ,Genome-wide association study ,Disease ,VARIANTS ,Biochemistry ,ANNOTATION ,0302 clinical medicine ,Phosphopantothenoylcysteine decarboxylase ,Single nucleotide ,Non-U.S. Gov't ,lcsh:Science ,skin and connective tissue diseases ,características del estudio::metaanálisis [CARACTERÍSTICAS DE PUBLICACIONES] ,Multidisciplinary ,Nucleid acid conformation ,integumentary system ,Research Support, Non-U.S. Gov't ,High-Throughput Nucleotide Sequencing ,Genètica - Tècnica ,3. Good health ,Nucleic acids ,Sequence analysis DNA ,Medical genetics ,medicine.medical_specialty ,Chromatin Immunoprecipitation ,GENES ,Investigative Techniques::Epidemiologic Methods::Epidemiologic Research Design::Genome-Wide Association Study [ANALYTICAL, DIAGNOSTIC AND THERAPEUTIC TECHNIQUES, AND EQUIPMENT] ,Science ,Non-P.H.S ,Single-nucleotide polymorphism ,Scleroderma systemic ,Computational biology ,Physics and Astronomy(all) ,Biology ,Research Support ,Study Characteristics::Meta-Analysis [PUBLICATION CHARACTERISTICS] ,Genetic polymorphisms ,enfermedades de la piel y tejido conjuntivo::enfermedades del tejido conjuntivo::esclerodermia sistémica [ENFERMEDADES] ,Polymorphism, Single Nucleotide ,General Biochemistry, Genetics and Molecular Biology ,Functional genomics, Genome-wide association studies, Systemic sclerosis, Rheumatic diseases ,CLASSIFICATION ,N.I.H ,03 medical and health sciences ,Research Support, N.I.H., Extramural ,Diacylglycerol kinase theta ,Molecular genetics ,REVEALS ,Journal Article ,medicine ,Humans ,Vascular Diseases ,Risk factor ,Polymorphism ,Genomes ,GENOME-WIDE ASSOCIATION ,METAANALYSIS ,030203 arthritis & rheumatology ,Scleroderma, Systemic ,Biochemistry, Genetics and Molecular Biology(all) ,Polimorfisme genètic ,HUMAN-CELLS ,Extramural ,Bayes Theorem ,General Chemistry ,Sequence Analysis, DNA ,Fibrosis ,030104 developmental biology ,Scleroderma (Disease) ,Skin and Connective Tissue Diseases::Connective Tissue Diseases::Scleroderma, Systemic [DISEASES] ,técnicas de investigación::métodos epidemiológicos::diseño de la investigación epidemiológica::estudio de asociación genómica completa [TÉCNICAS Y EQUIPOS ANALÍTICOS, DIAGNÓSTICOS Y TERAPÉUTICOS] ,Nucleic Acid Conformation ,VISUALIZATION ,lcsh:Q ,U.S. Gov't ,Esclerodèrmia ,Research Support, U.S. Gov't, Non-P.H.S ,Metaanàlisi ,Genetics and Molecular Biology(all) ,Genome-Wide Association Study - Abstract
Systemic sclerosis (SSc) is an autoimmune disease that shows one of the highest mortality rates among rheumatic diseases. We perform a large genome-wide association study (GWAS), and meta-analysis with previous GWASs, in 26,679 individuals and identify 27 independent genome-wide associated signals, including 13 new risk loci. The novel associations nearly double the number of genome-wide hits reported for SSc thus far. We define 95% credible sets of less than 5 likely causal variants in 12 loci. Additionally, we identify specific SSc subtype-associated signals. Functional analysis of high-priority variants shows the potential function of SSc signals, with the identification of 43 robust target genes through HiChIP. Our results point towards molecular pathways potentially involved in vasculopathy and fibrosis, two main hallmarks in SSc, and highlight the spectrum of critical cell types for the disease. This work supports a better understanding of the genetic basis of SSc and provides directions for future functional experiments., We thank Sofia Vargas, Sonia García, and Gema Robledo for their excellent technical assistance and all the patients and control donors for their essential collaboration. We thank National DNA Bank Carlos III (University of Salamanca, Spain) that supplied part of the control DNA samples from Spain, WTCCC and EIRA Consortiums, and PopGen 2.0 network. This work was supported by Spanish Ministry of Economy and Competitiveness (grant ref. SAF2015-66761-P), Consejeria de Innovacion, Ciencia y Tecnologia, Junta de Andalucía (P12-BIO-1395), Ministerio de Educación, Cultura y Deporte through the program FPU, Juan de la Cierva fellowship (FJCI-2015-24028), Red de Investigación en Inflamación y Enfermadades Reumaticas (RIER) from Instituto de Salud Carlos III (RD16/0012/0013), and Scleroderma Research Foundation and NIH P50- HG007735 (to H.Y.C.). H.Y.C. is an Investigator of the Howard Hughes Medical Institute. PopGen 2.0 is supported by a grant from the German Ministry for Education and Research (01EY1103). M.D.M and S.A. are supported by grant DoD W81XWH-18-1- 0423 and DoD W81XWH-16-1-0296, respectively
- Published
- 2019
- Full Text
- View/download PDF
3. BRAFV600 Mutations are Highly Consistent in Primary Melanomas and Matched Metastases- an Analysis of 160 Paired Tissue Samples by Real Time PCR and Next-Generation Sequencing: FC-010
- Author
-
Satzger, I, Marks, L., Klages, S., Kerick, M., Rüschoff, J., Middel, P., Kapp, A., Schacht, V., Völker, B., Timmermann, B., and Gutzmer, R.
- Published
- 2013
4. Erratum: Campylobacter jejuni impairs sodium transport and epithelial barrier function via cytokine release in human colon
- Author
-
Bücker, R., Krug, S.M., Moos, V., Bojarski, C., Schweiger, M.R., Kerick, M., Fromm, A., Janßen, S., Fromm, M., Hering, N.A., Siegmund, B., Schneider, T., Barmeyer, C., and Schulzke, J.D.
- Published
- 2018
- Full Text
- View/download PDF
5. Molecular stratification of autoimmune diseases based on epigenetic profiles
- Author
-
Barturen, G, Kerick, M, Alvarez-Errico, D, Quintares, R, Carnero, E, Gemperline, D, Dow, E, Beretta, Lorenzo, Pers, J.O., Renaudineau, Yves, Frostegard, J, Juárez, Manuela, Consortium, Clinical, Flow Cytometry, Group, Rao, S, Chamberlain, C, Wojcik, J., Segura, A, Martin, J, Ballestar, Esteban, Alarcon-Riquelme, Marta E., Centre for Genomics and Oncological Reearch (GENYO), Instituto de Parasitología y Biomedicina 'López-Neyra' (IPBLN), Consejo Superior de Investigaciones Científicas (CSIC), Institut d'Investigació Biomèdica de Bellvitge [Barcelone] (IDIBELL), Departmento de Quimica-Analitica, Granada, Eli Lilly, Indianapolis, USA, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, Lymphocyte B et Auto-immunité (LBAI), Université de Brest (UBO)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut Brestois Santé Agro Matière (IBSAM), Université de Brest (UBO), Laboratoire d'Immunologie et Immunothérapie [Brest], Centre Hospitalier Régional Universitaire de Brest (CHRU Brest), Unit of Chronic Diseases, Solna, Sweden, UCB Pharma Slough, Sanofi Genzyme, Quartzbio, Geneva, Departamento de Quimica-Analitica, Granada, Université de Brest (UBO)-Institut Brestois Santé Agro Matière (IBSAM), and Université de Brest (UBO)-Institut National de la Santé et de la Recherche Médicale (INSERM)
- Subjects
[SDV]Life Sciences [q-bio] ,ComputingMilieux_MISCELLANEOUS - Abstract
International audience
- Published
- 2018
6. Genomic and transcriptomic heterogeneity of colorectal tumors arising in Lynch Syndrome
- Author
-
Binder, H., Hopp, L., Schweiger, M., Hoffmann, S., Jühling, F., Kerick, M., Timmermann, B., Siebert, S., Grimm, C., Nersisyan, L., Arakelyan, A., Herberg, M., Buske, P., Loeffler-Wirth, H., Rosolowski, M., Engel, C., Przybilla, J., Peifer, M., Friedrichs, N., Moeslein, G., Odenthal, M., Hussong, M., Peters, S., Holzapfel, S., Nattermann, J., Hueneburg, R., Schmiegel, W., Royer-Pokora, B., Aretz, S., Kloth, M., Kloor, M., Buettner, R., Galle, J., and Loeffler, M.
- Published
- 2017
7. IDENTIFICATION OF NEW RISK LOCI AND PATHWAYS INVOLVED IN GCA PATHOGENESIS BY A GENOME-WIDE STUDY.
- Author
-
Borrego-Yaniz, G., Ortiz-Fernández, L., Kerick, M., Madrid-Paredes, A., Vaglio, A., Hernández-Rodríguez, J., Mackie, S., Castañeda, S., Solans-Laqué, R., Mestre, J., Dasgupta, B., Watts, R., Khalidi, N., Langford, C., Ytterberg, S. R., Beretta, L., Govoni, M., Emmi, G., Cimmino, M. A., and Witte, T.
- Published
- 2023
- Full Text
- View/download PDF
8. Somatic mutation profiles of MSI and MSS colorectal cancer identified by whole exome next generation sequencing and bioinformatics analysis
- Author
-
Timmermann, B., Kerick, M., Roehr, C., Fischer, A., Isau, M., Boerno, S., Wunderlich, A., Barmeyer, C., Seemann, P., Koenig, J., Lappe, M., Kuss, A., Garshasbi, M., Bertram, L., Trappe, K., Werber, M., Herrmann, B., Zatloukal, K., Lehrach, H., Schweiger, M., and This work was supported by the German Federal Ministry of Education and Research (01GS08105 'Mutanom,' 01GS08111 'Intestinal Modifers') and the Max Planck Society.
- Subjects
neoplasms ,digestive system diseases - Abstract
BACKGROUND: Colorectal cancer (CRC) is with approximately 1 million cases the third most common cancer worldwide. Extensive research is ongoing to decipher the underlying genetic patterns with the hope to improve early cancer diagnosis and treatment. In this direction, the recent progress in next generation sequencing technologies has revolutionized the field of cancer genomics. However, one caveat of these studies remains the large amount of genetic variations identified and their interpretation. METHODOLOGY/PRINCIPAL FINDINGS: Here we present the first work on whole exome NGS of primary colon cancers. We performed 454 whole exome pyrosequencing of tumor as well as adjacent not affected normal colonic tissue from microsatellite stable (MSS) and microsatellite instable (MSI) colon cancer patients and identified more than 50,000 small nucleotide variations for each tissue. According to predictions based on MSS and MSI pathomechanisms we identified eight times more somatic non-synonymous variations in MSI cancers than in MSS and we were able to reproduce the result in four additional CRCs. Our bioinformatics filtering approach narrowed down the rate of most significant mutations to 359 for MSI and 45 for MSS CRCs with predicted altered protein functions. In both CRCs, MSI and MSS, we found somatic mutations in the intracellular kinase domain of bone morphogenetic protein receptor 1A, BMPR1A, a gene where so far germline mutations are associated with juvenile polyposis syndrome, and show that the mutations functionally impair the protein function. CONCLUSIONS/SIGNIFICANCE: We conclude that with deep sequencing of tumor exomes one may be able to predict the microsatellite status of CRC and in addition identify potentially clinically relevant mutations.
- Published
- 2010
9. A SINGLE CELL TRANSCRIPTOMIC ANALYSIS REVEALS A PRO-INFLAMMATORY PROFILE IN PERIPHERAL BLOOD CD14+ MONOCYTES OF SYSTEMIC SCLEROSIS PATIENTS.
- Author
-
Villanueva-Martin, G., Acosta-Herrera, M., Carmona, E., Kerick, M., Ortego, N., Callejas-Rubio, J. L., Mages, N., Klages, S., Boerno, S., Timmermann, B., Bossini-Castillo, L., and Ibanez, J. Martin
- Published
- 2023
- Full Text
- View/download PDF
10. Microarray analysis of tumor necrosis factor alpha induced gene expression in U373 human glioblastoma cells
- Author
-
Schwamborn, J., Lindecke, A., Elvers, M., Horejschi, V., Kerick, M., Rafigh, M., Pfeiffer, J., Prullage, M., Kaltschmidt, Barbara, and Kaltschmidt, Christian
- Published
- 2003
11. Translation of next-generation sequencing (NGS) into molecular diagnostics.
- Author
-
Kotschote S, Wagner C, Marschall C, Mayer K, Hirv K, Kerick M, Timmermann B, and Klein H
- Abstract
In the past 5 years, next-generation sequencing (NGS) has been established as a valuable tool for several research applications. Commercially available platforms from Helicos, Illumina, Life Technologies, Pacific Biosciencies, and Roche allow for massively parallel sequencing and analysis in the fields of genomics, transcriptomics, and epigenomics. As in most projects, data throughput of the sequencers is not the limiting factor; genomic DNA samples are directly prepared for sequencing without prior conditioning. However, there are some applications such as targeted resequencing that do not require sequencing of whole genomes. Therefore, a technology called target enrichment was established more than 2 years ago. Different PCR- or hybridization-based approaches were further commercially developed and refined. The combination of this method with NGS can improve analysis of disease-related gene sets in molecular diagnostics by reducing time and costs. By taking advantage of the enormous data output, several genes and patients can be analyzed in parallel in one single instrument run. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
12. 836 STEROID HORMONES REGULATION OF METASTASIS- ASSOCIATED CHAPERONE PROTEINS AGR2 AND AGR3 IN PROSTATE CANCER CELLS
- Author
-
Bu, H., Schweiger, M., Fuchsberger, C., Wunderlich, A., Timmermann, B., Kerick, M., Manke, T., Cato, A.C.B., and Klocker, H.
- Published
- 2011
- Full Text
- View/download PDF
13. Identification of new risk loci shared across systemic vasculitides points towards potential target genes for drug repurposing
- Author
-
Ortiz-Fernández, Lourdes, Carmona, Elio G, Kerick, Martin, Lyons, Paul, Carmona, Francisco David, López Mejías, Raquel, Khor, Chiea Chuen, Grayson, Peter C, Tombetti, Enrico, Jiang, Lindi, Direskeneli, Haner, Saruhan-Direskeneli, Guher, Callejas-Rubio, José-Luis, Vaglio, Augusto, Salvarani, Carlo, Hernández-Rodríguez, Jose, Cid, Maria Cinta, Morgan, Ann W, Merkel, Peter A, Burgner, David, Smith, Kenneth Gc, Gonzalez-Gay, Miguel Angel, Sawalha, Amr H, Martin, Javier, Marquez, Ana, Ortiz-Fernández, Lourdes [0000-0002-0247-4280], Carmona, Francisco David [0000-0002-1427-7639], Grayson, Peter C [0000-0002-8269-9438], Salvarani, Carlo [0000-0003-3708-3148], Hernández-Rodríguez, Jose [0000-0002-2357-2015], Cid, Maria Cinta [0000-0002-4730-0938], Gonzalez-Gay, Miguel Angel [0000-0002-7924-7406], Martin, Javier [0000-0002-2202-0622], Marquez, Ana [0000-0001-9913-7688], Apollo - University of Cambridge Repository, and Ortiz-Fernandez L., Carmona E. G., Kerick M., Lyons P., Carmona F. D., Mejias R. L., Khor C. C., Grayson P. C., Tombetti E., Jiang L., et al.
- Subjects
Internal Diseases ,Vasculitis ,Immunology ,Autoimmunity ,SUSCEPTIBILITY ,VARIANTS ,Sağlık Bilimleri ,İmmünoloji ve Romatoloji ,İç Hastalıkları ,Clinical Medicine (MED) ,General Biochemistry, Genetics and Molecular Biology ,Immunology and Rheumatology ,ACTIVATION ,Genetic ,Rheumatology ,Health Sciences ,Humans ,Immunology and Allergy ,Klinik Tıp (MED) ,CTLA-4 Antigen ,Genetic Predisposition to Disease ,Polymorphism ,GENOME-WIDE ASSOCIATION ,ROMATOLOJİ ,Internal Medicine Sciences ,Polymorphism, Genetic ,Klinik Tıp ,Systemic Vasculitis ,Drug Repositioning ,Dahili Tıp Bilimleri ,CLINICAL MEDICINE ,Tıp ,Medicine ,Romatoloji ,Apoptosis Regulatory Proteins - Abstract
Objectives The number of susceptibility loci currently associated with vasculitis is lower than in other immune-mediated diseases due in part to small cohort sizes, a consequence of the low prevalence of vasculitides. This study aimed to identify new genetic risk loci for the main systemic vasculitides through a comprehensive analysis of their genetic overlap. Methods Genome-wide data from 8467 patients with any of the main forms of vasculitis and 29 795 healthy controls were meta-analysed using ASSET. Pleiotropic variants were functionally annotated and linked to their target genes. Prioritised genes were queried in DrugBank to identify potentially repositionable drugs for the treatment of vasculitis. Results Sixteen variants were independently associated with two or more vasculitides, 15 of them representing new shared risk loci. Two of these pleiotropic signals, located close to CTLA4 and CPLX1, emerged as novel genetic risk loci in vasculitis. Most of these polymorphisms appeared to affect vasculitis by regulating gene expression. In this regard, for some of these common signals, potential causal genes were prioritised based on functional annotation, including CTLA4, RNF145, IL12B, IL5, IRF1, IFNGR1, PTK2B, TRIM35, EGR2 and ETS2, each of which has key roles in inflammation. In addition, drug repositioning analysis showed that several drugs, including abatacept and ustekinumab, could be potentially repurposed in the management of the analysed vasculitides. Conclusions We identified new shared risk loci with functional impact in vasculitis and pinpointed potential causal genes, some of which could represent promising targets for the treatment of vasculitis., HELICAL Innovative Training Network, European Commission funded-under the Marie Sklodowska-Curie 813545, Cooperative Research Thematic Network programme RD16/0012/0013, Redes de Investigacion Cooperativa Orientadas a Resultados en Salud (RICORS) RD21/0002/0039, Instituto de Salud Carlos III PI18/00040, Juan de la Cierva Incorporacion fellowship IJC2019- 040746-I, United States Department of Health & Human Services, National Institutes of Health (NIH) - USA, NIH National Institute of Arthritis & Musculoskeletal & Skin Diseases (NIAMS) R01 AR070148, National Health and Medical Research Council (NHMRC) of Australia GTN1175744, Victorian Government's Operational Infrastructure Support Program, Rare Diseases Clinical Research Network (RDCRN), initiative of the Office of Rare Diseases Research (ORDR), NIH National Center for Advancing Translational Sciences (NCATS), NIH National Institute of Arthritis & Musculoskeletal & Skin Diseases (NIAMS) U54 AR057319, NIH National Center for Research Resources (NCRR) U54 RR019497
- Published
- 2023
14. The Effect of Body Fat Distribution on Systemic Sclerosis
- Author
-
Gonzalo Villanueva-Martin, Marialbert Acosta-Herrera, Martin Kerick, Elena López-Isac, Carmen P. Simeón, José L. Callejas, Shervin Assassi, Lorenzo Beretta, International SSc Group, Australian Scleroderma Interest Group (ASIG), Yannick Allanore, Susanna M. Proudman, Mandana Nikpour, Carmen Fonseca, Christopher P. Denton, Timothy R. D. J. Radstake, Maureen D. Mayes, Xia Jiang, Javier Martin, Lara Bossini-Castillo, Institut Català de la Salut, [Villanueva-Martin G, Kerick M, López-Isac E] Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Spain. [Acosta-Herrera M] Systemic Autoimmune Disease Unit, Hospital Clínico San Cecilio, Instituto de Investigación Biosanitaria Ibs, Granada, Spain. [Simeón CP] Servei de Medicina Interna, Vall d’Hebron Hospital Universitari, Barcelona, Spain. [Callejas JL] Department of Internal Medicine, Hospital San Cecilio, Granada, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
obesity ,systemic sclerosis ,mendelian randomization ,Skin and Connective Tissue Diseases::Connective Tissue Diseases::Scleroderma, Systemic [DISEASES] ,Esclerosi sistemàtica progressiva ,Obesitat ,Pathological Conditions, Signs and Symptoms::Signs and Symptoms::Body Weight::Overweight::Obesity [DISEASES] ,General Medicine ,afecciones patológicas, signos y síntomas::signos y síntomas::peso corporal::sobrepeso::obesidad [ENFERMEDADES] ,enfermedades de la piel y tejido conjuntivo::enfermedades del tejido conjuntivo::esclerodermia sistémica [ENFERMEDADES] - Abstract
Obesity contributes to a chronic proinflammatory state, which is a known risk factor to develop immune-mediated diseases. However, its role in systemic sclerosis (SSc) remains to be elucidated. Therefore, we conducted a two-sample mendelian randomization (2SMR) study to analyze the effect of three body fat distribution parameters in SSc. As instrumental variables, we used the allele effects described for single nucleotide polymorphisms (SNPs) in different genomewide association studies (GWAS) for SSc, body mass index (BMI), waist-to-hip ratio (WHR) and WHR adjusted for BMI (WHRadjBMI). We performed local (pHESS) and genome-wide (LDSC) genetic correlation analyses between each of the traits and SSc and we applied several Mendelian randomization (MR) methods (i.e., random effects inverse-variance weight, MR-Egger regression, MR pleiotropy residual sum and outlier method and a multivariable model). Our results show no genetic correlation or causal relationship between any of these traits and SSc. Nevertheless, we observed a negative causal association between WHRadjBMI and SSc, which might be due to the effect of gastrointestinal complications suffered by the majority of SSc patients. In conclusion, reverse causality might be an especially difficult confounding factor to define the effect of obesity in the onset of SSc., MCIN/AEI RTI2018101332-B-100 IJC2018-038026-I IJC2019-040080-I PRE2019-087586, "ERDF A way of making Europe" - European Union, Red de Investigacion en Inflamacion y Enfermedades Reumaticas (RIER) from Instituto de Salud Carlos III RD16/0012/0013, ESF Investing in your future
- Published
- 2022
15. Complement component C4 structural variation and quantitative traits contribute to sex-biased vulnerability in systemic sclerosis
- Author
-
Kerick, Martin, Acosta-Herrera, Marialbert, Simeón-Aznar, Carmen Pilar, Callejas, José Luis, Assassi, Shervin, International SSc Group, Proudman, Susanna M, Nikpour, Mandana, Australian Scleroderma Interest Group (ASIG), PRECISESADS Clinical Consortium, Hunzelmann, Nicolas, Moroncini, Gianluca, de Vries-Bouwstra, Jeska K, Orozco, Gisela, Barton, Anne, Herrick, Ariane L, Terao, Chikashi, Allanore, Yannick, Fonseca, Carmen, Alarcón-Riquelme, Marta Eugenia, Radstake, Timothy R D J, Beretta, Lorenzo, Denton, Christopher P, Mayes, Maureen D, Martin, Javier, Institut Català de la Salut, [Kerick M] Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Spain. [Acosta-Herrera M] Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Spain. Systemic Autoimmune Disease Unit, Hospital Clínico San Cecilio, Instituto de Investigación Biosanitaria Ibs. GRANADA, Granada, Spain. [Simeón-Aznar CP] Servei de Medicina Interna, Vall d’Hebron Hospital Universitari, Barcelona, Spain. [Callejas JL] Department of Internal Medicine, Hospital San Cecilio, Granada, Spain. [Assassi S] Department of Rheumatology, The University of Texas Health Science Center at Houston, Houston, TX, USA, Vall d'Hebron Barcelona Hospital Campus, Rheumatology, and AII - Inflammatory diseases
- Subjects
Otros calificadores::Otros calificadores::/genética [Otros calificadores] ,Skin and Connective Tissue Diseases::Connective Tissue Diseases::Scleroderma, Systemic [DISEASES] ,Genetics ,Other subheadings::Other subheadings::/genetics [Other subheadings] ,Inflammatory diseases Radboud Institute for Health Sciences [Radboudumc 5] ,Esclerosi sistemàtica progressiva - Aspectes genètics ,Genetic Phenomena::Genetic Phenomena::Genetic Structures::Transcriptome [PHENOMENA AND PROCESSES] ,Molecular Biology ,Expressió gènica ,enfermedades de la piel y tejido conjuntivo::enfermedades del tejido conjuntivo::esclerodermia sistémica [ENFERMEDADES] ,Genetics (clinical) ,fenómenos genéticos::expresión génica [FENÓMENOS Y PROCESOS] - Abstract
Copy number (CN) polymorphisms of complement C4 play distinct roles in many conditions, including immune-mediated diseases. We investigated the association of C4 CN with systemic sclerosis (SSc) risk. Imputed total C4, C4A, C4B, and HERV-K CN were analyzed in 26,633 individuals and validated in an independent cohort. Our results showed that higher C4 CN confers protection to SSc, and deviations from CN parity of C4A and C4B augmented risk. The protection contributed per copy of C4A and C4B differed by sex. Stronger protection was afforded by C4A in men and by C4B in women. C4 CN correlated well with its gene expression and serum protein levels, and less C4 was detected for both in SSc patients. Conditioned analysis suggests that C4 genetics strongly contributes to the SSc association within the major histocompatibility complex locus and highlights classical alleles and amino acid variants of HLA-DRB1 and HLA-DPB1 as C4-independent signals., MCIN/AEI by "ERDF A way of making Europe" RTI2018101332-B-100, Red de Investigacion en Inflamacion y Enfermedades Reumaticas (RIER) from Instituto de Salud Carlos III RD16/0012/0013, Innovative Medicines Initiative 1 & 2 Joint Undertaking (JU) 115565 831434, European Union's FP7 and Horizon 2020 research and innovation programs, EFPIA, Juan de la Cierva Incorporacion program - MCIN/AEI IJC2018-035131-I
- Published
- 2022
- Full Text
- View/download PDF
16. GWAS loci associated with Chagas cardiomyopathy influences DNA methylation levels
- Author
-
Desiré Casares-Marfil, Martin Kerick, Eduardo Andrés-León, Pau Bosch-Nicolau, Israel Molina, Chagas Genetics CYTED Network, Javier Martin, Marialbert Acosta-Herrera, Institut Català de la Salut, [Casares-Marfil D, Kerick M, Andrés-León E, Chagas Genetics CYTED Network] Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Spain. [Bosch-Nicolau P, Molina I] Unitat de Medicina Tropical i Salud Internacional, Vall d’Hebron Hospital Universitari, Barcelona, Spain. PROSICS, Barcelona, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
Chagas Cardiomyopathy ,Male ,Single Nucleotide Polymorphisms ,RC955-962 ,Genome-wide association study ,Cardiovascular Medicine ,Biochemistry ,fenómenos químicos::fenómenos bioquímicos::alquilación::metilación::metilación del ADN [FENÓMENOS Y PROCESOS] ,Medical Conditions ,Arctic medicine. Tropical medicine ,Medicine and Health Sciences ,Genetics ,DNA methylation ,Chemical Reactions ,ADN - Metilació ,Genomics ,Methylation ,Middle Aged ,Chemical Phenomena::Biochemical Phenomena::Alkylation::Methylation::DNA Methylation [PHENOMENA AND PROCESSES] ,Chromatin ,Nucleic acids ,Chemistry ,Infectious Diseases ,Cardiovascular Diseases ,Physical Sciences ,Phospholipases A2, Calcium-Independent ,Epigenetics ,Female ,Public aspects of medicine ,RA1-1270 ,Cardiomyopathies ,DNA modification ,Chromatin modification ,Research Article ,Neglected Tropical Diseases ,Chromosome biology ,Adult ,Cell biology ,Chagas, Malaltia de ,Investigative Techniques::Epidemiologic Methods::Epidemiologic Research Design::Genome-Wide Association Study [ANALYTICAL, DIAGNOSTIC AND THERAPEUTIC TECHNIQUES, AND EQUIPMENT] ,Quantitative Trait Loci ,Cardiology ,Miocardi - Malalties ,Single-nucleotide polymorphism ,Locus (genetics) ,Biology ,Quantitative trait locus ,Polymorphism, Single Nucleotide ,Parasitic Diseases::Protozoan Infections::Euglenozoa Infections::Trypanosomiasis::Chagas Disease::Chagas Cardiomyopathy [DISEASES] ,Parasitic Diseases ,Genome-Wide Association Studies ,Humans ,Chagas Disease ,Aged ,Genetic association ,Protozoan Infections ,Tumor Suppressor Proteins ,Public Health, Environmental and Occupational Health ,Biology and Life Sciences ,Computational Biology ,dNaM ,Human Genetics ,DNA ,enfermedades parasitarias::infecciones por protozoos::infecciones por Euglenozoa::tripanosomiasis::enfermedad de Chagas::miocardiopatía chagásica [ENFERMEDADES] ,Tropical Diseases ,Genome Analysis ,DNA Polymerase I ,Repressor Proteins ,Genetic Loci ,Genetics of Disease ,técnicas de investigación::métodos epidemiológicos::diseño de la investigación epidemiológica::estudio de asociación genómica completa [TÉCNICAS Y EQUIPOS ANALÍTICOS, DIAGNÓSTICOS Y TERAPÉUTICOS] ,Gene expression ,Carrier Proteins ,Genome-Wide Association Study - Abstract
A recent genome-wide association study (GWAS) identified a locus in chromosome 11 associated with the chronic cardiac form of Chagas disease. Here we aimed to elucidate the potential functional mechanism underlying this genetic association by analyzing the correlation among single nucleotide polymorphisms (SNPs) and DNA methylation (DNAm) levels as cis methylation quantitative trait loci (cis-mQTL) within this region. A total of 2,611 SNPs were tested against 2,647 DNAm sites, in a subset of 37 chronic Chagas cardiomyopathy patients and 20 asymptomatic individuals from the GWAS. We identified 6,958 significant cis-mQTLs (False Discovery Rate [FDR], Author summary Genome-wide association studies (GWAS) have provided extensive information regarding the genetic component of complex traits, including parasitic diseases such as Chagas disease. However, these associations mapped in regulatory regions of the genome and assigning them a functional consequence have been cumbersome. In this study we aimed to evaluate the functional mechanism underlying the previously reported genomic association with chronic Chagas cardiomyopathy, by assessing the correlation between methylation changes and the underlying genetic variations within the region. These methylation quantitative trait loci (mQTLs) may be involved in gene expression regulation. We identified mQTLs in three genes that have been associated with cardiovascular diseases in previous studies. Interestingly, one of these genes was previously identified as differentially methylated and expressed in heart biopsies of chronic Chagas cardiomyopathy patients. Our results suggest novel genes that could play a role in the chronic Chagas cardiomyopathy, evidencing the functional relevance of the previously associated loci.
- Published
- 2021
17. Comprehensive analysis of the major histocompatibility complex in systemic sclerosis identifies differential HLA associations by clinical and serological subtypes
- Author
-
Acosta-Herrera, Marialbert, Kerick, Martin, Lopéz-Isac, Elena, Assassi, Shervin, Beretta, Lorenzo, Simeón-Aznar, Carmen Pilar, Ortego-Centeno, Norberto, International SSc Group, Proudman, Susanna M, Australian Scleroderma Interest Group (ASIG), Hunzelmann, Nicolas, Moroncini, Gianluca, de Vries-Bouwstra, Jeska K, Orozco, Gisela, Barton, Anne, Herrick, Ariane L, Terao, Chikashi, Allanore, Yannick, Brown, Matthew A, Radstake, Timothy Rdj, Fonseca, Carmen, Denton, Christopher P, Mayes, Maureen D, Martin, Javier, Ministerio de Ciencia e Innovación (España), Instituto de Salud Carlos III, National Institutes of Health (US), Manchester Biomedical Research Centre, Universidad de Cantabria, Institut Català de la Salut, [Acosta-Herrera M, Kerick M, Lopéz-Isac E] Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Andalucía, Spain. [Assassi S] Rheumatology and Clinical Immunogenetics, The University of Texas Health Science Center at Houston, Houston, Texas, USA. [Beretta L] Referral Center for Systemic Autoimmune Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy. [Simeón-Aznar CP] Servei de Medicina Interna, Vall d’Hebron Hospital Universitari, Barcelona, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
0301 basic medicine ,autoantibodies ,systemic sclerosis ,Immunology ,Locus (genetics) ,Single-nucleotide polymorphism ,Human leukocyte antigen ,Major histocompatibility complex ,General Biochemistry, Genetics and Molecular Biology ,polymorphism ,Major Histocompatibility Complex ,03 medical and health sciences ,0302 clinical medicine ,Antígens HLA ,Rheumatology ,Polymorphism (computer science) ,Other subheadings::Other subheadings::/immunology [Other subheadings] ,Immunology and Allergy ,Medicine ,Humans ,Genetic Predisposition to Disease ,Otros calificadores::Otros calificadores::/inmunología [Otros calificadores] ,Allele ,skin and connective tissue diseases ,Alleles ,030203 arthritis & rheumatology ,Skin and Connective Tissue Diseases::Skin and Connective Tissue Diseases::Skin Diseases::Scleroderma, Systemic [DISEASES] ,Scleroderma, Systemic ,Amino Acids, Peptides, and Proteins::Proteins::Glycoproteins::Membrane Glycoproteins::Histocompatibility Antigens Class II [CHEMICALS AND DRUGS] ,biology ,immune complex diseases ,business.industry ,enfermedades de la piel y tejido conjuntivo::enfermedades de la piel y tejido conjuntivo::enfermedades de la piel::esclerodermia sistémica [ENFERMEDADES] ,Autoantibody ,030104 developmental biology ,biology.protein ,Esclerosi sistemàtica progressiva ,genetic ,Immunogenètica ,business ,Immune complex disease ,aminoácidos, péptidos y proteínas::proteínas::glicoproteínas::glicoproteínas de membranas::antígenos de histocompatibilidad de clase II [COMPUESTOS QUÍMICOS Y DROGAS] ,Genome-Wide Association Study ,HLA-DRB1 Chains - Abstract
Objective The greatest genetic effect reported for systemic sclerosis (SSc) lies in the major histocompatibility complex (MHC) locus. Leveraging the largest SSc genome-wide association study, we aimed to fine-map this region to identify novel human leucocyte antigen (HLA) genetic variants associated with SSc susceptibility and its main clinical and serological subtypes. Methods 9095 patients with SSc and 17 584 controls genome-wide genotyped were used to impute and test single-nucleotide polymorphisms (SNPs) across the MHC, classical HLA alleles and their composite amino acid residues. Additionally, patients were stratified according to their clinical and serological status, namely, limited cutaneous systemic sclerosis (lcSSc), diffuse cutaneous systemic sclerosis (dcSSc), anticentromere (ACA), antitopoisomerase (ATA) and anti-RNApolIII autoantibodies (ARA). Results Sequential conditional analyses showed nine SNPs, nine classical alleles and seven amino acids that modelled the observed associations with SSc. This confirmed previously reported associations with HLA-DRB1*11:04 and HLA-DPB1*13:01, and revealed a novel association of HLA-B*08:01. Stratified analyses showed specific associations of HLA-DQA1*02:01 with lcSSc, and an exclusive association of HLA-DQA1*05:01 with dcSSc. Similarly, private associations were detected in HLA-DRB1*08:01 and confirmed the previously reported association of HLA-DRB1*07:01 with ACA-positive patients, as opposed to the HLA-DPA1*02:01 and HLA-DQB1*03:01 alleles associated with ATA presentation. Conclusions This study confirms the contribution of HLA class II and reveals a novel association of HLA class I with SSc, suggesting novel pathways of disease pathogenesis. Furthermore, we describe specific HLA associations with SSc clinical and serological subtypes that could serve as biomarkers of disease severity and progression., This work was supported by the Spanish Ministry of Science and Innovation (grant ref. SAF2015-66761-P and RTI20181013 (32-B-100)), Red de Investigación en Inflamación y Enfermedades Reumáticas from Instituto de Salud Carlos III (RD16/0012/0013) and grants from National Institutes of Health (R01AR073284) and DoD (W81XWH-16-1-0296). MAH was funded by the Spanish Ministry of Science and Innovation through the Juan de la Cierva Incorporacion program (ref. IJC2018-035131-I). GO, AB and ALH were supported by the NIHR Manchester Biomedical Research Centre and Versus Arthritis (grant ref 21754)
- Published
- 2021
18. Cross-trait GWAS in COVID-19 and systemic sclerosis reveals novel genes implicated in fibrotic and inflammation pathways.
- Author
-
Rosa-Baez C, Borrego-Yaniz G, Rodriguez-Martin I, Kerick M, Acosta-Herrera M, Martín J, and Ortiz-Fernández L
- Abstract
Objectives: COVID-19 and systemic sclerosis (SSc) share multiple similarities in their clinical manifestations, alterations in immune response, and therapeutic options. These resemblances have also been identified in other immune-mediated inflammatory diseases where a common genetic component has been found. Thus, we decided to evaluate for the first time this shared genetic architecture with SSc., Methods: For this study, we retrieved genomic data from two European-ancestry cohorts: 2,597 856 individuals from The COVID-19 Host Genetics Initiative consortium, and 26 679 individuals from the largest genomic scan in SSc. We performed a cross-trait meta-analyses including >9.3 million SNPs. Finally, we conducted functional annotation to prioritize potential causal genes and performed drug repurposing analysis., Results: Our results revealed a total of 19 non-HLA pleiotropic loci, including 2 novel associations for both conditions (BMP1 and PPARG), and 12 emerging as new shared loci. Functional annotation of these regions underscored their potential regulatory role and identified potential causal genes, many of which are implicated in fibrotic and inflammatory pathways. Remarkably, we observed an antagonistic pleiotropy model of the IFN signalling between COVID-19 and SSc, including the well-known TYK2 P1104A missense variant, showing a protective effect for SSc while being a risk factor for COVID-19, along with two additional novel pleiotropic associations (IRF8 and SENP7). Finally, our findings provide new therapeutic options that could potentially benefit both conditions., Conclusion: Our study confirms the genetic resemblance between susceptibility to and severity of COVID-19 and SSc, revealing a novel common genetic contribution affecting fibrotic and immune pathways., (© The Author(s) 2025. Published by Oxford University Press on behalf of the British Society for Rheumatology.)
- Published
- 2025
- Full Text
- View/download PDF
19. A strong Dysregulated Myeloid Component in the Epigenetic Landscape of Systemic Sclerosis: An Integrated DNA Methylome and Transcriptome Analysis.
- Author
-
Martínez-López J, Ortiz-Fernandez L, Estupiñán-Moreno E, Kerick M, Andrés-León E, Terron-Camero LC, Carnero-Montoro E, Barturen G, Beretta L, Almeida I, Alarcón-Riquelme ME, Ballestar E, Acosta-Herrera M, and Martín J
- Abstract
Objective: Nongenetic factors influence systemic sclerosis (SSc) pathogenesis, underscoring epigenetics as a relevant contributor to the disease. We aimed to unravel DNA methylation abnormalities associated with SSc through an epigenome-wide association study., Methods: We analyzed DNA methylation data from whole-blood samples in 179 patients with SSc and 241 unaffected individuals to identify differentially methylated positions (DMPs) with a false discovery rate (FDR) <0.05. These results were further integrated with RNA sequencing data from the same patients to assess their functional consequence. Additionally, we examined the impact of DNA methylation changes on transcription factors and analyzed the relationship between alterations of the methylation and gene expression profile and serum proteins levels., Results: This analysis yielded 525 DMPs enriched in immune-related pathways, with leukocyte cell-cell adhesion being the most significant (FDR = 4.91 × 10
-9 ), prioritizing integrins as they were exposed by integrating methylome and transcriptome data. Furthermore, through this integrative approach, we observed an enrichment of neutrophil-related pathways, highlighting this myeloid cell type as a relevant contributor in SSc pathogenesis. In addition, we uncovered novel profibrotic and proinflammatory mechanisms involved in the disease. Finally, the altered epigenetic and transcriptomic signature revealed an increased activity of CCAAT/enhancer-binding protein transcription factor family in SSc, which is crucial in the myeloid lineage development., Conclusion: Our findings uncover the impaired epigenetic regulation of the disease and its impact on gene expression, identifying new molecules for potential clinical applications and improving our understanding of SSc pathogenesis., (© 2024 The Author(s). Arthritis & Rheumatology published by Wiley Periodicals LLC on behalf of American College of Rheumatology.)- Published
- 2024
- Full Text
- View/download PDF
20. Stratification according to autoantibody status in systemic sclerosis reveals distinct molecular signatures.
- Author
-
Rouvière B, Le Dantec C, Bettacchioli E, Beretta L, Foulquier N, Cao C, Jamin C, Pers JO, Kerick M, Martin J, Alarcón-Riquelme ME, de Moreuil C, Cornec D, and Hillion S
- Abstract
Objectives: Systemic sclerosis (SSc) is a heterogeneous disease, complicating its management. Its complexity and the insufficiency of clinical manifestations alone to delineate homogeneous patient groups further challenge this task. However, autoantibodies could serve as relevant markers for the pathophysiological mechanisms driving the disease. Identifying specific immunological mechanisms based on patients' serological statuses might facilitate a deeper understanding of the diversity of the disease., Methods: A cohort of 206 patients with SSc enrolled in the PRECISESADS cross-sectional study was examined. Patients were stratified based on their anti-centromere (ACA) and anti-SCL70 (SCL70) antibody statuses. Comprehensive omics analyses including transcriptomic, flow cytometric, cytokine and metabolomic data were analysed to characterise the differences between these patient groups., Results: Patients with SCL70 antibodies showed severe clinical features such as diffuse cutaneous sclerosis and pulmonary fibrosis and were biologically distinguished by unique transcriptomic profiles. They exhibit a pro-inflammatory and fibrotic signature associated with impaired tissue remodelling and increased carnitine metabolism. Conversely, ACA-positive patients exhibited an immunomodulation and tissue homeostasis signature and increased phospholipid metabolism., Conclusions: Patients with SSc display varying biological profiles based on their serological status. The findings highlight the potential utility of serological status as a discriminating factor in disease severity and suggest its relevance in tailoring treatment strategies and future research directions., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2024. Re-use permitted under CC BY. Published by BMJ on behalf of EULAR.)
- Published
- 2024
- Full Text
- View/download PDF
21. Molecular subtypes explain lupus epigenomic heterogeneity unveiling new regulatory genetic risk variants.
- Author
-
Castellini-Pérez O, Povedano E, Barturen G, Martínez-Bueno M, Iakovliev A, Kerick M, López-Domínguez R, Marañón C, Martín J, Ballestar E, Borghi MO, Qiu W, Zhu C, Shankara S, Spiliopoulou A, de Rinaldis E, Carnero-Montoro E, and Alarcón-Riquelme ME
- Abstract
The heterogeneity of systemic lupus erythematosus (SLE) can be explained by epigenetic alterations that disrupt transcriptional programs mediating environmental and genetic risk. This study evaluated the epigenetic contribution to SLE heterogeneity considering molecular and serological subtypes, genetics and transcriptional status, followed by drug target discovery. We performed a stratified epigenome-wide association studies of whole blood DNA methylation from 213 SLE patients and 221 controls. Methylation quantitative trait loci analyses, cytokine and transcription factor activity - epigenetic associations and methylation-expression correlations were conducted. New drug targets were searched for based on differentially methylated genes. In a stratified approach, a total of 974 differential methylation CpG sites with dependency on molecular subtypes and autoantibody profiles were found. Mediation analyses suggested that SLE-associated SNPs in the HLA region exert their risk through DNA methylation changes. Novel genetic variants regulating DNAm in disease or in specific molecular contexts were identified. The epigenetic landscapes showed strong association with transcription factor activity and cytokine levels, conditioned by the molecular context. Epigenetic signals were enriched in known and novel drug targets for SLE. This study reveals possible genetic drivers and consequences of epigenetic variability on SLE heterogeneity and disentangles the DNAm mediation role on SLE genetic risk and novel disease-specific meQTLs. Finally, novel targets for drug development were discovered., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
22. Risk loci involved in giant cell arteritis susceptibility: a genome-wide association study.
- Author
-
Borrego-Yaniz G, Ortiz-Fernández L, Madrid-Paredes A, Kerick M, Hernández-Rodríguez J, Mackie SL, Vaglio A, Castañeda S, Solans R, Mestre-Torres J, Khalidi N, Langford CA, Ytterberg S, Beretta L, Govoni M, Emmi G, Cimmino MA, Witte T, Neumann T, Holle J, Schönau V, Pugnet G, Papo T, Haroche J, Mahr A, Mouthon L, Molberg Ø, Diamantopoulos AP, Voskuyl A, Daikeler T, Berger CT, Molloy ES, Blockmans D, van Sleen Y, Iles M, Sorensen L, Luqmani R, Reynolds G, Bukhari M, Bhagat S, Ortego-Centeno N, Brouwer E, Lamprecht P, Klapa S, Salvarani C, Merkel PA, Cid MC, González-Gay MA, Morgan AW, Martin J, and Márquez A
- Subjects
- Humans, Genetic Loci genetics, Female, Male, Aged, Polymorphism, Single Nucleotide, Middle Aged, Case-Control Studies, Giant Cell Arteritis genetics, Giant Cell Arteritis pathology, Genome-Wide Association Study, Genetic Predisposition to Disease
- Abstract
Background: Giant cell arteritis is an age-related vasculitis that mainly affects the aorta and its branches in individuals aged 50 years and older. Current options for diagnosis and treatment are scarce, highlighting the need to better understand its underlying pathogenesis. Genome-wide association studies (GWAS) have emerged as a powerful tool for unravelling the pathogenic mechanisms involved in complex diseases. We aimed to characterise the genetic basis of giant cell arteritis by performing the largest GWAS of this vasculitis to date and to assess the functional consequences and clinical implications of identified risk loci., Methods: We collected and meta-analysed genomic data from patients with giant cell arteritis and healthy controls of European ancestry from ten cohorts across Europe and North America. Eligible patients required confirmation of giant cell arteritis diagnosis by positive temporal artery biopsy, positive temporal artery doppler ultrasonography, or imaging techniques confirming large-vessel vasculitis. We assessed the functional consequences of loci associated with giant cell arteritis using cell enrichment analysis, fine-mapping, and causal gene prioritisation. We also performed a drug repurposing analysis and developed a polygenic risk score to explore the clinical implications of our findings., Findings: We included a total of 3498 patients with giant cell arteritis and 15 550 controls. We identified three novel loci associated with risk of giant cell arteritis. Two loci, MFGE8 (rs8029053; p=4·96 × 10
-8 ; OR 1·19 [95% CI 1·12-1·26]) and VTN (rs704; p=2·75 × 10-9 ; OR 0·84 [0·79-0·89]), were related to angiogenesis pathways and the third locus, CCDC25 (rs11782624; p=1·28 × 10-8 ; OR 1·18 [1·12-1·25]), was related to neutrophil extracellular traps (NETs). We also found an association between this vasculitis and HLA region and PLG. Variants associated with giant cell arteritis seemed to fulfil a specific regulatory role in crucial immune cell types. Furthermore, we identified several drugs that could represent promising candidates for treatment of this disease. The polygenic risk score model was able to identify individuals at increased risk of developing giant cell arteritis (90th percentile OR 2·87 [95% CI 2·15-3·82]; p=1·73 × 10-13 )., Interpretation: We have found several additional loci associated with giant cell arteritis, highlighting the crucial role of angiogenesis in disease susceptibility. Our study represents a step forward in the translation of genomic findings to clinical practice in giant cell arteritis, proposing new treatments and a method to measure genetic predisposition to this vasculitis., Funding: Institute of Health Carlos III, Spanish Ministry of Science and Innovation, UK Medical Research Council, and National Institute for Health and Care Research., Competing Interests: Declaration of interests MCC reports support from the Spanish Ministry of Science and Innovation (PID2020-114909RB-I00), Vasculitis Foundation, Agency for the Management of University and Research Grants (2021 SGR 01561), and Kiniksa Pharmaceuticals; consulting fees or honoraria from GSK, CSL Vifor, AbbVie, and AstraZeneca; support for attending meetings from Kiniksa Pharmaceuticals; and participation on a data safety monitoring board or advisory board for GSK, CSL Vifor, and AstraZeneca. GE has acted as a consultant for GSK, AstraZeneca, Sobi, Novartis, Boehringer, and CSL Vifor. AWM reports support from the UK Medical Research Council (MRC), National Institute for Health and Care Research (NIHR), Leeds Care, and Roche Products; and consulting fees or honoraria from CSL Vifor and AstraZeneca. PL reports grants or contracts from the Federal Ministry of Education and Research, German Research Society, German Society for Rheumatology, John Grube Foundation, and CSL Vifor; consulting fees or honoraria from GSK, CSL Vifor, AstraZeneca, Bristol Myers Squibb, Boehringer Ingelheim, Forum für medizinische Fortbildung, Janssen, Rheumaakademie, and UCB; support for attending meetings from CSL Vifor; and participation on a data safety monitoring board or advisory board for GSK, CSL Vifor, AbbVie, and Novartis. TW reports consulting fees or honoraria from AbbVie, AstraZeneca, Lilly, UCB, and Novartis; and participation on a data safety monitoring board or advisory board for AbbVie, AstraZeneca, Lilly, UCB, Novartis, and Fresenius. MAG-G reports honoraria from GSK. NK reports grants or contracts from Bristol Myers Squibb, AbbVie, and Sanofi; and consulting fees or honoraria from Roche, Otsuka, GSK, and Mallinckrodt. CAL reports grants or contracts from Bristol Myers Squibb and support from the National Institutes of Health. PAM reports grants, contracts, or consulting fees from AbbVie, Amgen, AstraZeneca, ArGenx, Boehringer Ingelheim, Bristol Myers Squibb, Cabaletta, CSL Behring, Eicos, Electra, Forbius, Genentech–Roche, GSK, HiBio, InflaRx, Janssen, Jubilant, Kyverna, MiroBio, Neutrolis, Novartis, NS Pharma, Q32, Regeneron, Sanofi, Sparrow, Takeda, and Vistera; royalties or licenses from UpToDate; and stock or stock options from Kyverna, Q32, and Sparrow. SLM reports grants or contracts from MRC, NIHR, and CSL Vifor; consulting fees from Roche, Sanofi, AbbVie, AstraZeneca, and Pfizer; payment or honoraria for lectures or educational events from Roche, Pfizer, UCB, CSL Vifor, Fresenius Kabi, and Novartis; support for attending meetings from Pfizer; participation on a data safety monitoring board or advisory board for Collaboration for Leadership in Applied Health Research and Care, Haywood Foundation, and GC-SheaLD; a leadership or fiduciary role in the British Society for Rheumatology Clinical Affairs Committee; participation as an investigator on industry-sponsored clinical trials for Sanofi; and infrastructure support from MRC. LB reports payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing, or educational events from Instrumentation Laboratory SPA and AbbVie; and support for attending meetings from AbbVie and Novartis. EB reports payments or honoraria from EULAR and received grants from the Dutch Arthritis Society DAS and the EU/EFPIA/Innovative Medicines Initiative 2 Joint Undertaking Immune-Image grant no 831514. EB is member of the board of the non-profit organisation, Auto-immune Research Hub, in the Netherlands. All other authors declare no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY 4.0 license. Published by Elsevier Ltd.. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
23. Genome-Wide Association Study Identifies the First Germline Genetic Variant Associated With Erdheim-Chester Disease.
- Author
-
Martínez-López J, Márquez A, Pegoraro F, Ortiz-Fernández L, Acosta-Herrera M, Kerick M, Gelain E, Diamond EL, Durham BH, Abdel-Wahab O, Go RS, Koster MJ, Dagna L, Campochiaro C, Collin M, Milne P, Estrada-Veras JI, O'Brien K, Papo M, Cohen-Aubar F, Amoura Z, Haroche J, Martín J, and Vaglio A
- Subjects
- Humans, Genome-Wide Association Study, Genomics, Germ Cells pathology, Erdheim-Chester Disease genetics, Erdheim-Chester Disease pathology
- Abstract
Objective: Erdheim-Chester disease (ECD) is rare histiocytosis with a wide range of clinical manifestations. Somatic mutations are key to the pathogenesis of the disease; however, the relationship between germline genetic variants and ECD has not been examined so far. The present study aims to explore the inherited genetic component of ECD by performing the first genome-wide association study., Methods: After quality controls, a cohort of 255 patients with ECD and 7,471 healthy donors was included in this study. Afterward, a logistic regression followed by in silico functional annotation was performed., Results: A signal at the 18q12.3 genomic region was identified as a new susceptibility locus for ECD (P = 2.75 × 10
-11 ; Odds Ratio = 2.09). This association was annotated to the SETBP1 gene, which is involved in clonal haematopoiesis. Functional annotation of this region and of the identified suggestive signals revealed additional genes that could be potentially involved in the pathogenesis of the disease., Conclusion: Overall, this work demonstrates that germline genetic variants can impact on the development of ECD and suggests new pathways with a potential pathogenic role., (© 2023 The Authors. Arthritis & Rheumatology published by Wiley Periodicals LLC on behalf of American College of Rheumatology.)- Published
- 2024
- Full Text
- View/download PDF
24. A holistic approach to understanding immune-mediated inflammatory diseases: bioinformatic tools to integrate omics data.
- Author
-
Borrego-Yaniz G, Terrón-Camero LC, Kerick M, Andrés-León E, and Martin J
- Abstract
Immune-mediated inflammatory diseases (IMIDs) comprise a complex group of pathologies with diverse etiologies and clinical manifestations. In particular, omics technologies have remodeled our understanding of a set of IMIDs such as systemic autoimmune rheumatic diseases (SARDs), generating vast amounts of data on the genome, epigenome, transcriptome, proteome and metabolome of immune cells and SARDs patients. However, the integration of omics data to advance our knowledge of these diseases is challenging, requiring advanced bioinformatic tools. This review explores different multi-omic integrative tools for refining previous research, exploring the biological relevance of datasets within different contexts, or translating omics results into clinical advances. We also discuss relevant multi-omic studies in SARDs research and the potential of omics data from available repositories to complement ongoing investigation in this field., Competing Interests: The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (© 2023 The Authors.)
- Published
- 2023
- Full Text
- View/download PDF
25. Non-classical circulating monocytes expressing high levels of microsomal prostaglandin E2 synthase-1 tag an aberrant IFN-response in systemic sclerosis.
- Author
-
Villanueva-Martin G, Acosta-Herrera M, Carmona EG, Kerick M, Ortego-Centeno N, Callejas-Rubio JL, Mages N, Klages S, Börno S, Timmermann B, Bossini-Castillo L, and Martin J
- Subjects
- Humans, Female, Male, Biomarkers, Middle Aged, Adult, Gene Expression Profiling, Single-Cell Analysis, Aged, Transcriptome, Lipopolysaccharide Receptors metabolism, Intramolecular Oxidoreductases metabolism, Intramolecular Oxidoreductases genetics, Scleroderma, Systemic immunology, Scleroderma, Systemic metabolism, Scleroderma, Systemic pathology, Monocytes immunology, Monocytes metabolism, Interferons metabolism
- Abstract
Systemic sclerosis (SSc) is a complex disease that affects the connective tissue, causing fibrosis. SSc patients show altered immune cell composition and activation in the peripheral blood (PB). PB monocytes (Mos) are recruited into tissues where they differentiate into macrophages, which are directly involved in fibrosis. To understand the role of CD14
+ PB Mos in SSc, a single-cell transcriptome analysis (scRNA-seq) was conducted on 8 SSc patients and 8 controls. Using unsupervised clustering methods, CD14+ cells were assigned to 11 clusters, which added granularity to the known monocyte subsets: classical (cMos), intermediate (iMos) and non-classical Mos (ncMos) or type 2 dendritic cells. NcMos were significantly overrepresented in SSc patients and showed an active IFN-signature and increased expression levels of PTGES, in addition to monocyte motility and adhesion markers. We identified a SSc-related cluster of IRF7+ STAT1+ iMos with an aberrant IFN-response. Finally, a depletion of M2 polarised cMos in SSc was observed. Our results highlighted the potential of PB Mos as biomarkers for SSc and provided new possibilities for putative drug targets for modulating the innate immune response in SSc., (Copyright © 2023 The Authors. Published by Elsevier Ltd.. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
26. Identification of new risk loci shared across systemic vasculitides points towards potential target genes for drug repurposing.
- Author
-
Ortiz-Fernández L, Carmona EG, Kerick M, Lyons P, Carmona FD, López Mejías R, Khor CC, Grayson PC, Tombetti E, Jiang L, Direskeneli H, Saruhan-Direskeneli G, Callejas-Rubio JL, Vaglio A, Salvarani C, Hernández-Rodríguez J, Cid MC, Morgan AW, Merkel PA, Burgner D, Smith KG, Gonzalez-Gay MA, Sawalha AH, Martin J, and Marquez A
- Subjects
- Humans, CTLA-4 Antigen, Drug Repositioning, Genetic Predisposition to Disease genetics, Apoptosis Regulatory Proteins genetics, Systemic Vasculitis genetics, Vasculitis drug therapy, Vasculitis genetics
- Abstract
Objectives: The number of susceptibility loci currently associated with vasculitis is lower than in other immune-mediated diseases due in part to small cohort sizes, a consequence of the low prevalence of vasculitides. This study aimed to identify new genetic risk loci for the main systemic vasculitides through a comprehensive analysis of their genetic overlap., Methods: Genome-wide data from 8467 patients with any of the main forms of vasculitis and 29 795 healthy controls were meta-analysed using ASSET. Pleiotropic variants were functionally annotated and linked to their target genes. Prioritised genes were queried in DrugBank to identify potentially repositionable drugs for the treatment of vasculitis., Results: Sixteen variants were independently associated with two or more vasculitides, 15 of them representing new shared risk loci. Two of these pleiotropic signals, located close to CTLA4 and CPLX1 , emerged as novel genetic risk loci in vasculitis. Most of these polymorphisms appeared to affect vasculitis by regulating gene expression. In this regard, for some of these common signals, potential causal genes were prioritised based on functional annotation, including CTLA4 , RNF145 , IL12B , IL5 , IRF1 , IFNGR1 , PTK2B , TRIM35 , EGR2 and ETS2 , each of which has key roles in inflammation. In addition, drug repositioning analysis showed that several drugs, including abatacept and ustekinumab, could be potentially repurposed in the management of the analysed vasculitides., Conclusions: We identified new shared risk loci with functional impact in vasculitis and pinpointed potential causal genes, some of which could represent promising targets for the treatment of vasculitis., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2023
- Full Text
- View/download PDF
27. Identification of Mechanisms by Which Genetic Susceptibility Loci Influence Systemic Sclerosis Risk Using Functional Genomics in Primary T Cells and Monocytes.
- Author
-
González-Serna D, Shi C, Kerick M, Hankinson J, Ding J, McGovern A, Tutino M, Villanueva-Martin G, Ortego-Centeno N, Callejas JL, Martin J, and Orozco G
- Subjects
- Humans, Genetic Predisposition to Disease genetics, Genetic Loci, Genomics, Monocytes, Scleroderma, Systemic pathology
- Abstract
Objective: Systemic sclerosis (SSc) is a complex autoimmune disease with a strong genetic component. However, most of the genes associated with the disease are still unknown because associated variants affect mostly noncoding intergenic elements of the genome. We used functional genomics to translate the genetic findings into a better understanding of the disease., Methods: Promoter capture Hi-C and RNA-sequencing experiments were performed in CD4+ T cells and CD14+ monocytes from 10 SSc patients and 5 healthy controls to link SSc-associated variants with their target genes, followed by differential expression and differential interaction analyses between cell types., Results: We linked SSc-associated loci to 39 new potential target genes and confirmed 7 previously known SSc-associated genes. We highlight novel causal genes, such as CXCR5, as the most probable candidate gene for the DDX6 locus. Some previously known SSc-associated genes, such as IRF8, STAT4, and CD247, showed cell type-specific interactions. We also identified 15 potential drug targets already in use in other similar immune-mediated diseases that could be repurposed for SSc treatment. Furthermore, we observed that interactions were directly correlated with the expression of important genes implicated in cell type-specific pathways and found evidence that chromatin conformation is associated with genotype., Conclusion: Our study revealed potential causal genes for SSc-associated loci, some of them acting in a cell type-specific manner, suggesting novel biologic mechanisms that might mediate SSc pathogenesis., (© 2022 The Authors. Arthritis & Rheumatology published by Wiley Periodicals LLC on behalf of American College of Rheumatology.)
- Published
- 2023
- Full Text
- View/download PDF
28. FLT3 functional low-frequency variant rs76428106-C is associated with susceptibility to systemic sclerosis.
- Author
-
Martínez-López J, Kerick M, Ortiz-Fernández L, Acosta-Herrera M, Márquez A, and Martín J
- Subjects
- Humans, Genetic Predisposition to Disease, Genome-Wide Association Study, Polymorphism, Single Nucleotide, Genotype, Case-Control Studies, fms-Like Tyrosine Kinase 3 genetics, Scleroderma, Systemic genetics, Autoimmune Diseases genetics
- Abstract
Objectives: rs76428106-C, a low frequency polymorphism that affects the splicing of the FLT3 gene, has recently been associated with several seropositive autoimmune diseases. Here, we aimed to evaluate the potential implication of rs76428106-C in the susceptibility to systemic sclerosis (SSc)., Methods: We analysed a total of 26 598 European ancestry individuals, 9063 SSc and 17 535 healthy controls, to test the association between FLT3 rs76428106-C and SSc and its different subphenotypes. Genotype data of rs76428106 were obtained by imputation of already available genome-wide association study data and analysed by logistic regression analysis., Results: In accordance with that observed in other autoimmune disorders, the FLT3 rs76428106-C allele was significantly increased [P-value = 2.03 × 10-3, odds ratio (OR) = 1.34] in SSc patients compared with healthy controls. A similar risk effect was found when the main SSc clinical and serological subgroups were compared with controls. When comparing SSc patients with and without digital ulcers (DU), the rs76428106-C frequency was significantly increased in DU-positive SSc patients in comparison with DU-negative patients (P-value = 0.036, OR = 2.16)., Conclusion: This study is the first to report an association between rs76428176-C and SSc. Our results support the role of FLT3 as a relevant gene in seropositive immune-mediated diseases and a potential biomarker for SSc microangiopathy., (© The Author(s) 2022. Published by Oxford University Press on behalf of the British Society for Rheumatology.)
- Published
- 2023
- Full Text
- View/download PDF
29. Multi-ancestry genome-wide association analyses identify novel genetic mechanisms in rheumatoid arthritis.
- Author
-
Ishigaki K, Sakaue S, Terao C, Luo Y, Sonehara K, Yamaguchi K, Amariuta T, Too CL, Laufer VA, Scott IC, Viatte S, Takahashi M, Ohmura K, Murasawa A, Hashimoto M, Ito H, Hammoudeh M, Emadi SA, Masri BK, Halabi H, Badsha H, Uthman IW, Wu X, Lin L, Li T, Plant D, Barton A, Orozco G, Verstappen SMM, Bowes J, MacGregor AJ, Honda S, Koido M, Tomizuka K, Kamatani Y, Tanaka H, Tanaka E, Suzuki A, Maeda Y, Yamamoto K, Miyawaki S, Xie G, Zhang J, Amos CI, Keystone E, Wolbink G, van der Horst-Bruinsma I, Cui J, Liao KP, Carroll RJ, Lee HS, Bang SY, Siminovitch KA, de Vries N, Alfredsson L, Rantapää-Dahlqvist S, Karlson EW, Bae SC, Kimberly RP, Edberg JC, Mariette X, Huizinga T, Dieudé P, Schneider M, Kerick M, Denny JC, Matsuda K, Matsuo K, Mimori T, Matsuda F, Fujio K, Tanaka Y, Kumanogoh A, Traylor M, Lewis CM, Eyre S, Xu H, Saxena R, Arayssi T, Kochi Y, Ikari K, Harigai M, Gregersen PK, Yamamoto K, Louis Bridges S Jr, Padyukov L, Martin J, Klareskog L, Okada Y, and Raychaudhuri S
- Subjects
- Humans, Genetic Predisposition to Disease, Polymorphism, Single Nucleotide genetics, Asian People genetics, Adaptor Proteins, Signal Transducing genetics, Genome-Wide Association Study, Arthritis, Rheumatoid genetics
- Abstract
Rheumatoid arthritis (RA) is a highly heritable complex disease with unknown etiology. Multi-ancestry genetic research of RA promises to improve power to detect genetic signals, fine-mapping resolution and performances of polygenic risk scores (PRS). Here, we present a large-scale genome-wide association study (GWAS) of RA, which includes 276,020 samples from five ancestral groups. We conducted a multi-ancestry meta-analysis and identified 124 loci (P < 5 × 10
-8 ), of which 34 are novel. Candidate genes at the novel loci suggest essential roles of the immune system (for example, TNIP2 and TNFRSF11A) and joint tissues (for example, WISP1) in RA etiology. Multi-ancestry fine-mapping identified putatively causal variants with biological insights (for example, LEF1). Moreover, PRS based on multi-ancestry GWAS outperformed PRS based on single-ancestry GWAS and had comparable performance between populations of European and East Asian ancestries. Our study provides several insights into the etiology of RA and improves the genetic predictability of RA., (© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2022
- Full Text
- View/download PDF
30. The Effect of Body Fat Distribution on Systemic Sclerosis.
- Author
-
Villanueva-Martin G, Acosta-Herrera M, Kerick M, López-Isac E, Simeón CP, Callejas JL, Assassi S, Beretta L, SSc Group I, Asig ASIG, Allanore Y, Proudman SM, Nikpour M, Fonseca C, Denton CP, Radstake TRDJ, Mayes MD, Jiang X, Martin J, and Bossini-Castillo L
- Abstract
Obesity contributes to a chronic proinflammatory state, which is a known risk factor to develop immune-mediated diseases. However, its role in systemic sclerosis (SSc) remains to be elucidated. Therefore, we conducted a two-sample mendelian randomization (2SMR) study to analyze the effect of three body fat distribution parameters in SSc. As instrumental variables, we used the allele effects described for single nucleotide polymorphisms (SNPs) in different genome-wide association studies (GWAS) for SSc, body mass index (BMI), waist-to-hip ratio (WHR) and WHR adjusted for BMI (WHRadjBMI). We performed local (pHESS) and genome-wide (LDSC) genetic correlation analyses between each of the traits and SSc and we applied several Mendelian randomization (MR) methods (i.e., random effects inverse-variance weight, MR-Egger regression, MR pleiotropy residual sum and outlier method and a multivariable model). Our results show no genetic correlation or causal relationship between any of these traits and SSc. Nevertheless, we observed a negative causal association between WHRadjBMI and SSc, which might be due to the effect of gastrointestinal complications suffered by the majority of SSc patients. In conclusion, reverse causality might be an especially difficult confounding factor to define the effect of obesity in the onset of SSc.
- Published
- 2022
- Full Text
- View/download PDF
31. Integrative epigenomics in Sjögren´s syndrome reveals novel pathways and a strong interaction between the HLA, autoantibodies and the interferon signature.
- Author
-
Teruel M, Barturen G, Martínez-Bueno M, Castellini-Pérez O, Barroso-Gil M, Povedano E, Kerick M, Català-Moll F, Makowska Z, Buttgereit A, Pers JO, Marañón C, Ballestar E, Martin J, Carnero-Montoro E, and Alarcón-Riquelme ME
- Subjects
- DNA Methylation genetics, Female, Humans, Male, Sjogren's Syndrome etiology, Autoantibodies, Epigenomics, Gene Expression genetics, Gene Expression Regulation genetics, Genetic Variation, HLA Antigens genetics, Interferons genetics, Sjogren's Syndrome genetics, Sjogren's Syndrome immunology
- Abstract
Primary Sjögren's syndrome (SS) is a systemic autoimmune disease characterized by lymphocytic infiltration and damage of exocrine salivary and lacrimal glands. The etiology of SS is complex with environmental triggers and genetic factors involved. By conducting an integrated multi-omics study, we confirmed a vast coordinated hypomethylation and overexpression effects in IFN-related genes, what is known as the IFN signature. Stratified and conditional analyses suggest a strong interaction between SS-associated HLA genetic variation and the presence of Anti-Ro/SSA autoantibodies in driving the IFN epigenetic signature and determining SS. We report a novel epigenetic signature characterized by increased DNA methylation levels in a large number of genes enriched in pathways such as collagen metabolism and extracellular matrix organization. We identified potential new genetic variants associated with SS that might mediate their risk by altering DNA methylation or gene expression patterns, as well as disease-interacting genetic variants that exhibit regulatory function only in the SS population. Our study sheds new light on the interaction between genetics, autoantibody profiles, DNA methylation and gene expression in SS, and contributes to elucidate the genetic architecture of gene regulation in an autoimmune population., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
32. Admixture mapping analysis reveals differential genetic ancestry associated with Chagas disease susceptibility in the Colombian population.
- Author
-
Casares-Marfil D, Guillen-Guio B, Lorenzo-Salazar JM, Rodríguez-Pérez H, Kerick M, Jaimes-Campos MA, Díaz ML, Estupiñán E, Echeverría LE, González CI, Martín J, Flores C, and Acosta-Herrera M
- Subjects
- Colombia, Disease Susceptibility, Hispanic or Latino, Humans, Chagas Disease genetics, Polymorphism, Single Nucleotide genetics
- Abstract
Chagas disease is an infection caused by the parasite Trypanosoma cruzi, endemic in Latino America. Leveraging the three-way admixture between Native American (AMR), European (EUR) and African (AFR) populations in Latin Americans, we aimed to better understand the genetic basis of Chagas disease by performing an admixture mapping study in a Colombian population. A two-stage study was conducted, and subjects were classified as seropositive and seronegative for T. cruzi. In stage 1, global and local ancestries were estimated using reference data from the 1000 Genomes Project (1KGP), and local ancestry associations were performed by logistic regression models. The AMR ancestry showed a protective association with Chagas disease within the major histocompatibility complex region [Odds ratio (OR) = 0.74, 95% confidence interval (CI) = 0.66-0.83, lowest P-value = 4.53 × 10-8]. The fine mapping assessment on imputed genotypes combining data from stage 1 and 2 from an independent Colombian cohort, revealed nominally associated variants in high linkage disequilibrium with the top signal (rs2032134, OR = 0.93, 95% CI = 0.90-0.97, P-value = 3.54 × 10-4) in the previously associated locus. To assess ancestry-specific adaptive signals, a selective sweep scan in an AMR reference population from 1KGP together with an in silico functional analysis highlighted the Tripartite Motif family and the human leukocyte antigen genes, with crucial role in the immune response against pathogens. Furthermore, these analyses emphasized the macrophages, neutrophils and eosinophils, as key players in the defense against T. cruzi. This first admixture mapping study in Chagas disease provided novel insights underlying the host immune response in the pathogenesis of this neglected disease., (© The Author(s) 2021. Published by Oxford University Press.)
- Published
- 2021
- Full Text
- View/download PDF
33. The metabolic growth limitations of petite cells lacking the mitochondrial genome.
- Author
-
Vowinckel J, Hartl J, Marx H, Kerick M, Runggatscher K, Keller MA, Mülleder M, Day J, Weber M, Rinnerthaler M, Yu JSL, Aulakh SK, Lehmann A, Mattanovich D, Timmermann B, Zhang N, Dunn CD, MacRae JI, Breitenbach M, and Ralser M
- Subjects
- Amino Acids metabolism, Biomass, Cell Proliferation, Citric Acid Cycle, Fungal Proteins chemistry, Fungal Proteins genetics, Fungal Proteins metabolism, Membrane Potential, Mitochondrial, Mutation, Phenotype, Structure-Activity Relationship, Energy Metabolism, Genome, Mitochondrial, Mitochondria genetics, Mitochondria metabolism, Yeasts genetics, Yeasts metabolism
- Abstract
Eukaryotic cells can survive the loss of their mitochondrial genome, but consequently suffer from severe growth defects. 'Petite yeasts', characterized by mitochondrial genome loss, are instrumental for studying mitochondrial function and physiology. However, the molecular cause of their reduced growth rate remains an open question. Here we show that petite cells suffer from an insufficient capacity to synthesize glutamate, glutamine, leucine and arginine, negatively impacting their growth. Using a combination of molecular genetics and omics approaches, we demonstrate the evolution of fast growth overcomes these amino acid deficiencies, by alleviating a perturbation in mitochondrial iron metabolism and by restoring a defect in the mitochondrial tricarboxylic acid cycle, caused by aconitase inhibition. Our results hence explain the slow growth of mitochondrial genome-deficient cells with a partial auxotrophy in four amino acids that results from distorted iron metabolism and an inhibited tricarboxylic acid cycle., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2021
- Full Text
- View/download PDF
34. GWAS loci associated with Chagas cardiomyopathy influences DNA methylation levels.
- Author
-
Casares-Marfil D, Kerick M, Andrés-León E, Bosch-Nicolau P, Molina I, Martin J, and Acosta-Herrera M
- Subjects
- Adult, Aged, Carrier Proteins genetics, Carrier Proteins metabolism, Chagas Cardiomyopathy metabolism, DNA Methylation, DNA Polymerase I genetics, DNA Polymerase I metabolism, Female, Genome-Wide Association Study, Humans, Male, Middle Aged, Phospholipases A2, Calcium-Independent genetics, Phospholipases A2, Calcium-Independent metabolism, Polymorphism, Single Nucleotide, Quantitative Trait Loci, Repressor Proteins genetics, Repressor Proteins metabolism, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins metabolism, Chagas Cardiomyopathy genetics
- Abstract
A recent genome-wide association study (GWAS) identified a locus in chromosome 11 associated with the chronic cardiac form of Chagas disease. Here we aimed to elucidate the potential functional mechanism underlying this genetic association by analyzing the correlation among single nucleotide polymorphisms (SNPs) and DNA methylation (DNAm) levels as cis methylation quantitative trait loci (cis-mQTL) within this region. A total of 2,611 SNPs were tested against 2,647 DNAm sites, in a subset of 37 chronic Chagas cardiomyopathy patients and 20 asymptomatic individuals from the GWAS. We identified 6,958 significant cis-mQTLs (False Discovery Rate [FDR]<0.05) at 1 Mb each side of the GWAS leading variant, where six of them potentially modulate the expression of the SAC3D1 gene, the reported gene in the previous GWAS. In addition, a total of 268 cis-mQTLs showed differential methylation between chronic Chagas cardiomyopathy patients and asymptomatic individuals. The most significant cis-mQTLs mapped in the gene bodies of POLA2 (FDR = 1.04x10-11), PLAAT3 (FDR = 7.22x10-03), and CCDC88B (FDR = 1.89x10-02) that have been associated with cardiovascular and hematological traits in previous studies. One of the most relevant interactions correlated with hypermethylation of CCDC88B. This gene is involved in the inflammatory response, and its methylation and expression levels have been previously reported in Chagas cardiomyopathy. Our findings support the functional relevance of the previously associated genomic region, highlighting the regulation of novel genes that could play a role in the chronic cardiac form of the disease., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2021
- Full Text
- View/download PDF
35. Genomic and phenotypic insights from an atlas of genetic effects on DNA methylation.
- Author
-
Min JL, Hemani G, Hannon E, Dekkers KF, Castillo-Fernandez J, Luijk R, Carnero-Montoro E, Lawson DJ, Burrows K, Suderman M, Bretherick AD, Richardson TG, Klughammer J, Iotchkova V, Sharp G, Al Khleifat A, Shatunov A, Iacoangeli A, McArdle WL, Ho KM, Kumar A, Söderhäll C, Soriano-Tárraga C, Giralt-Steinhauer E, Kazmi N, Mason D, McRae AF, Corcoran DL, Sugden K, Kasela S, Cardona A, Day FR, Cugliari G, Viberti C, Guarrera S, Lerro M, Gupta R, Bollepalli S, Mandaviya P, Zeng Y, Clarke TK, Walker RM, Schmoll V, Czamara D, Ruiz-Arenas C, Rezwan FI, Marioni RE, Lin T, Awaloff Y, Germain M, Aïssi D, Zwamborn R, van Eijk K, Dekker A, van Dongen J, Hottenga JJ, Willemsen G, Xu CJ, Barturen G, Català-Moll F, Kerick M, Wang C, Melton P, Elliott HR, Shin J, Bernard M, Yet I, Smart M, Gorrie-Stone T, Shaw C, Al Chalabi A, Ring SM, Pershagen G, Melén E, Jiménez-Conde J, Roquer J, Lawlor DA, Wright J, Martin NG, Montgomery GW, Moffitt TE, Poulton R, Esko T, Milani L, Metspalu A, Perry JRB, Ong KK, Wareham NJ, Matullo G, Sacerdote C, Panico S, Caspi A, Arseneault L, Gagnon F, Ollikainen M, Kaprio J, Felix JF, Rivadeneira F, Tiemeier H, van IJzendoorn MH, Uitterlinden AG, Jaddoe VWV, Haley C, McIntosh AM, Evans KL, Murray A, Räikkönen K, Lahti J, Nohr EA, Sørensen TIA, Hansen T, Morgen CS, Binder EB, Lucae S, Gonzalez JR, Bustamante M, Sunyer J, Holloway JW, Karmaus W, Zhang H, Deary IJ, Wray NR, Starr JM, Beekman M, van Heemst D, Slagboom PE, Morange PE, Trégouët DA, Veldink JH, Davies GE, de Geus EJC, Boomsma DI, Vonk JM, Brunekreef B, Koppelman GH, Alarcón-Riquelme ME, Huang RC, Pennell CE, van Meurs J, Ikram MA, Hughes AD, Tillin T, Chaturvedi N, Pausova Z, Paus T, Spector TD, Kumari M, Schalkwyk LC, Visscher PM, Davey Smith G, Bock C, Gaunt TR, Bell JT, Heijmans BT, Mill J, and Relton CL
- Subjects
- Chromosome Mapping, Epigenesis, Genetic genetics, Genome-Wide Association Study, Humans, Multifactorial Inheritance genetics, Polymorphism, Single Nucleotide genetics, Quantitative Trait, Heritable, Transcriptome genetics, DNA metabolism, DNA Methylation genetics, Gene Expression Regulation genetics, Genetic Predisposition to Disease genetics, Quantitative Trait Loci genetics
- Abstract
Characterizing genetic influences on DNA methylation (DNAm) provides an opportunity to understand mechanisms underpinning gene regulation and disease. In the present study, we describe results of DNAm quantitative trait locus (mQTL) analyses on 32,851 participants, identifying genetic variants associated with DNAm at 420,509 DNAm sites in blood. We present a database of >270,000 independent mQTLs, of which 8.5% comprise long-range (trans) associations. Identified mQTL associations explain 15-17% of the additive genetic variance of DNAm. We show that the genetic architecture of DNAm levels is highly polygenic. Using shared genetic control between distal DNAm sites, we constructed networks, identifying 405 discrete genomic communities enriched for genomic annotations and complex traits. Shared genetic variants are associated with both DNAm levels and complex diseases, but only in a minority of cases do these associations reflect causal relationships from DNAm to trait or vice versa, indicating a more complex genotype-phenotype map than previously anticipated., (© 2021. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2021
- Full Text
- View/download PDF
36. Comprehensive analysis of the major histocompatibility complex in systemic sclerosis identifies differential HLA associations by clinical and serological subtypes.
- Author
-
Acosta-Herrera M, Kerick M, Lopéz-Isac E, Assassi S, Beretta L, Simeón-Aznar CP, Ortego-Centeno N, Proudman SM, Hunzelmann N, Moroncini G, de Vries-Bouwstra JK, Orozco G, Barton A, Herrick AL, Terao C, Allanore Y, Brown MA, Radstake TR, Fonseca C, Denton CP, Mayes MD, and Martin J
- Subjects
- Alleles, Genetic Predisposition to Disease, HLA-DRB1 Chains genetics, Humans, Major Histocompatibility Complex, Genome-Wide Association Study, Scleroderma, Systemic genetics
- Abstract
Objective: The greatest genetic effect reported for systemic sclerosis (SSc) lies in the major histocompatibility complex (MHC) locus. Leveraging the largest SSc genome-wide association study, we aimed to fine-map this region to identify novel human leucocyte antigen (HLA) genetic variants associated with SSc susceptibility and its main clinical and serological subtypes., Methods: 9095 patients with SSc and 17 584 controls genome-wide genotyped were used to impute and test single-nucleotide polymorphisms (SNPs) across the MHC, classical HLA alleles and their composite amino acid residues. Additionally, patients were stratified according to their clinical and serological status, namely, limited cutaneous systemic sclerosis (lcSSc), diffuse cutaneous systemic sclerosis (dcSSc), anticentromere (ACA), antitopoisomerase (ATA) and anti-RNApolIII autoantibodies (ARA)., Results: Sequential conditional analyses showed nine SNPs, nine classical alleles and seven amino acids that modelled the observed associations with SSc. This confirmed previously reported associations with HLA-DRB1*11:04 and HLA-DPB1*13:01 , and revealed a novel association of HLA-B*08:01 . Stratified analyses showed specific associations of HLA-DQA1*02:01 with lcSSc, and an exclusive association of HLA-DQA1*05:01 with dcSSc. Similarly, private associations were detected in HLA-DRB1*08:01 and confirmed the previously reported association of HLA-DRB1*07:01 with ACA-positive patients, as opposed to the HLA-DPA1*02:01 and HLA-DQB1*03:01 alleles associated with ATA presentation., Conclusions: This study confirms the contribution of HLA class II and reveals a novel association of HLA class I with SSc, suggesting novel pathways of disease pathogenesis. Furthermore, we describe specific HLA associations with SSc clinical and serological subtypes that could serve as biomarkers of disease severity and progression., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2021
- Full Text
- View/download PDF
37. Expression Quantitative Trait Locus Analysis in Systemic Sclerosis Identifies New Candidate Genes Associated With Multiple Aspects of Disease Pathology.
- Author
-
Kerick M, González-Serna D, Carnero-Montoro E, Teruel M, Acosta-Herrera M, Makowska Z, Buttgereit A, Babaei S, Barturen G, López-Isac E, Lesche R, Beretta L, Alarcon-Riquelme ME, and Martin J
- Subjects
- Adult, Aged, Basic Helix-Loop-Helix Transcription Factors genetics, Female, Genetic Association Studies, Humans, Inhibitor of Differentiation Proteins genetics, Kruppel-Like Factor 4, Kruppel-Like Transcription Factors genetics, Male, Middle Aged, Molecular Targeted Therapy, Nuclear Proteins genetics, Polymorphism, Single Nucleotide, Quantitative Trait Loci, T-Box Domain Proteins genetics, Transcription Factors genetics, Gene Expression Regulation genetics, Scleroderma, Systemic genetics
- Abstract
Objective: To identify the genetic variants that affect gene expression (expression quantitative trait loci [eQTLs]) in systemic sclerosis (SSc) and to investigate their role in the pathogenesis of the disease., Methods: We performed an eQTL analysis using whole-blood sequencing data from 333 SSc patients and 524 controls and integrated them with SSc genome-wide association study (GWAS) data. We integrated our findings from expression modeling, differential expression analysis, and transcription factor binding site enrichment with key clinical features of SSc., Results: We detected 49,123 validated cis-eQTLs from 4,539 SSc-associated single-nucleotide polymorphisms (SNPs) (P
GWAS < 10-5 ). A total of 1,436 genes were within 1 Mb of the 4,539 SSc-associated SNPs. Of those 1,436 genes, 565 were detected as having ≥1 eQTL with an SSc-associated SNP. We developed a strategy to prioritize disease-associated genes based on their expression variance explained by SSc eQTLs (r2 > 0.05). As a result, 233 candidates were identified, 134 (58%) of them associated with hallmarks of SSc and 105 (45%) of them differentially expressed in the blood cells, skin, or lung tissue of SSc patients. Transcription factor binding site analysis revealed enriched motifs of 24 transcription factors (5%) among SSc eQTLs, 5 of which were found to be differentially regulated in the blood cells (ELF1 and MGA), skin (KLF4 and ID4), and lungs (TBX4) of SSc patients. Ten candidate genes (4%) can be targeted by approved medications for immune-mediated diseases, of which only 3 have been tested in clinical trials in patients with SSc., Conclusion: The findings of the present study indicate a new layer to the molecular complexity of SSc, contributing to a better understanding of the pathogenesis of the disease., (© 2021, American College of Rheumatology.)- Published
- 2021
- Full Text
- View/download PDF
38. Integrative Analysis Reveals a Molecular Stratification of Systemic Autoimmune Diseases.
- Author
-
Barturen G, Babaei S, Català-Moll F, Martínez-Bueno M, Makowska Z, Martorell-Marugán J, Carmona-Sáez P, Toro-Domínguez D, Carnero-Montoro E, Teruel M, Kerick M, Acosta-Herrera M, Le Lann L, Jamin C, Rodríguez-Ubreva J, García-Gómez A, Kageyama J, Buttgereit A, Hayat S, Mueller J, Lesche R, Hernandez-Fuentes M, Juarez M, Rowley T, White I, Marañón C, Gomes Anjos T, Varela N, Aguilar-Quesada R, Garrancho FJ, López-Berrio A, Rodriguez Maresca M, Navarro-Linares H, Almeida I, Azevedo N, Brandão M, Campar A, Faria R, Farinha F, Marinho A, Neves E, Tavares A, Vasconcelos C, Trombetta E, Montanelli G, Vigone B, Alvarez-Errico D, Li T, Thiagaran D, Blanco Alonso R, Corrales Martínez A, Genre F, López Mejías R, Gonzalez-Gay MA, Remuzgo S, Ubilla Garcia B, Cervera R, Espinosa G, Rodríguez-Pintó I, De Langhe E, Cremer J, Lories R, Belz D, Hunzelmann N, Baerlecken N, Kniesch K, Witte T, Lehner M, Stummvoll G, Zauner M, Aguirre-Zamorano MA, Barbarroja N, Castro-Villegas MC, Collantes-Estevez E, de Ramon E, Díaz Quintero I, Escudero-Contreras A, Fernández Roldán MC, Jiménez Gómez Y, Jiménez Moleón I, Lopez-Pedrera R, Ortega-Castro R, Ortego N, Raya E, Artusi C, Gerosa M, Meroni PL, Schioppo T, De Groof A, Ducreux J, Lauwerys B, Maudoux AL, Cornec D, Devauchelle-Pensec V, Jousse-Joulin S, Jouve PE, Rouvière B, Saraux A, Simon Q, Alvarez M, Chizzolini C, Dufour A, Wynar D, Balog A, Bocskai M, Deák M, Dulic S, Kádár G, Kovács L, Cheng Q, Gerl V, Hiepe F, Khodadadi L, Thiel S, de Rinaldis E, Rao S, Benschop RJ, Chamberlain C, Dow ER, Ioannou Y, Laigle L, Marovac J, Wojcik J, Renaudineau Y, Borghi MO, Frostegård J, Martín J, Beretta L, Ballestar E, McDonald F, Pers JO, and Alarcón-Riquelme ME
- Subjects
- Adult, Aged, Antiphospholipid Syndrome genetics, Antiphospholipid Syndrome immunology, Arthritis, Rheumatoid genetics, Arthritis, Rheumatoid immunology, Autoimmune Diseases immunology, Case-Control Studies, Cluster Analysis, Cross-Sectional Studies, Epigenomics, Female, Humans, Inflammation immunology, Interferons immunology, Lupus Erythematosus, Systemic genetics, Lupus Erythematosus, Systemic immunology, Male, Middle Aged, Mixed Connective Tissue Disease genetics, Mixed Connective Tissue Disease immunology, Scleroderma, Systemic genetics, Scleroderma, Systemic immunology, Sjogren's Syndrome genetics, Sjogren's Syndrome immunology, Undifferentiated Connective Tissue Diseases genetics, Undifferentiated Connective Tissue Diseases immunology, Autoimmune Diseases classification, Autoimmune Diseases genetics, Epigenome, Gene Expression Profiling
- Abstract
Objective: Clinical heterogeneity, a hallmark of systemic autoimmune diseases, impedes early diagnosis and effective treatment, issues that may be addressed if patients could be classified into groups defined by molecular pattern. This study was undertaken to identify molecular clusters for reclassifying systemic autoimmune diseases independently of clinical diagnosis., Methods: Unsupervised clustering of integrated whole blood transcriptome and methylome cross-sectional data on 955 patients with 7 systemic autoimmune diseases and 267 healthy controls was undertaken. In addition, an inception cohort was prospectively followed up for 6 or 14 months to validate the results and analyze whether or not cluster assignment changed over time., Results: Four clusters were identified and validated. Three were pathologic, representing "inflammatory," "lymphoid," and "interferon" patterns. Each included all diagnoses and was defined by genetic, clinical, serologic, and cellular features. A fourth cluster with no specific molecular pattern was associated with low disease activity and included healthy controls. A longitudinal and independent inception cohort showed a relapse-remission pattern, where patients remained in their pathologic cluster, moving only to the healthy one, thus showing that the molecular clusters remained stable over time and that single pathogenic molecular signatures characterized each individual patient., Conclusion: Patients with systemic autoimmune diseases can be jointly stratified into 3 stable disease clusters with specific molecular patterns differentiating different molecular disease mechanisms. These results have important implications for future clinical trials and the study of nonresponse to therapy, marking a paradigm shift in our view of systemic autoimmune diseases., (© 2020, American College of Rheumatology.)
- Published
- 2021
- Full Text
- View/download PDF
39. Combined analysis of transcriptomic and genetic data for the identification of loci involved in glucocorticosteroid response in asthma.
- Author
-
Hernandez-Pacheco N, Gorenjak M, Jurgec S, Corrales A, Jorgensen A, Karimi L, Vijverberg SJ, Berce V, Schieck M, Acosta-Herrera M, Kerick M, Samedy-Bates LA, Tavendale R, Villar J, Mukhopadhyay S, Pirmohamed M, Verhamme KMC, Kabesch M, Hawcutt DB, Turner S, Palmer CN, Burchard EG, Maitland-van der Zee AH, Flores C, Potočnik U, and Pino-Yanes M
- Subjects
- Genome-Wide Association Study, Humans, Polymorphism, Single Nucleotide, Asthma drug therapy, Asthma genetics, Transcriptome
- Published
- 2021
- Full Text
- View/download PDF
40. Genomic Risk Score impact on susceptibility to systemic sclerosis.
- Author
-
Bossini-Castillo L, Villanueva-Martin G, Kerick M, Acosta-Herrera M, López-Isac E, Simeón CP, Ortego-Centeno N, Assassi S, Hunzelmann N, Gabrielli A, de Vries-Bouwstra JK, Allanore Y, Fonseca C, Denton CP, Radstake TR, Alarcón-Riquelme ME, Beretta L, Mayes MD, and Martin J
- Subjects
- Adult, Aged, Antibodies, Antinuclear immunology, Arthritis, Rheumatoid genetics, Autoantibodies immunology, Case-Control Studies, DNA Topoisomerases immunology, Female, Genetic Predisposition to Disease, Genome-Wide Association Study, Humans, Linear Models, Lupus Erythematosus, Systemic genetics, Male, Middle Aged, Polymorphism, Single Nucleotide, Risk Factors, Scleroderma, Diffuse immunology, Scleroderma, Limited immunology, Scleroderma, Systemic genetics, Scleroderma, Systemic immunology, Sjogren's Syndrome genetics, White People, Scleroderma, Diffuse genetics, Scleroderma, Limited genetics
- Abstract
Objectives: Genomic Risk Scores (GRS) successfully demonstrated the ability of genetics to identify those individuals at high risk for complex traits including immune-mediated inflammatory diseases (IMIDs). We aimed to test the performance of GRS in the prediction of risk for systemic sclerosis (SSc) for the first time., Methods: Allelic effects were obtained from the largest SSc Genome-Wide Association Study (GWAS) to date (9 095 SSc and 17 584 healthy controls with European ancestry). The best-fitting GRS was identified under the additive model in an independent cohort that comprised 400 patients with SSc and 571 controls. Additionally, GRS for clinical subtypes (limited cutaneous SSc and diffuse cutaneous SSc) and serological subtypes (anti-topoisomerase positive (ATA+) and anti-centromere positive (ACA+)) were generated. We combined the estimated GRS with demographic and immunological parameters in a multivariate generalised linear model., Results: The best-fitting SSc GRS included 33 single nucleotide polymorphisms (SNPs) and discriminated between patients with SSc and controls (area under the receiver operating characteristic (ROC) curve (AUC)=0.673). Moreover, the GRS differentiated between SSc and other IMIDs, such as rheumatoid arthritis and Sjögren's syndrome. Finally, the combination of GRS with age and immune cell counts significantly increased the performance of the model (AUC=0.787). While the SSc GRS was not able to discriminate between ATA+ and ACA+ patients (AUC<0.5), the serological subtype GRS, which was based on the allelic effects observed for the comparison between ACA+ and ATA+ patients, reached an AUC=0.693., Conclusions: GRS was successfully implemented in SSc. The model discriminated between patients with SSc and controls or other IMIDs, confirming the potential of GRS to support early and differential diagnosis for SSc., Competing Interests: Competing interests: LB-C: none; GV-M: none; MK: none; MA-H: none; ELI: none; International SSc Group: none; PRECISESADS Consortium: none; MEAl-R: none; LB: none; JM: none., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2021
- Full Text
- View/download PDF
41. Genome-wide whole blood transcriptome profiling in a large European cohort of systemic sclerosis patients.
- Author
-
Beretta L, Barturen G, Vigone B, Bellocchi C, Hunzelmann N, De Langhe E, Cervera R, Gerosa M, Kovács L, Ortega Castro R, Almeida I, Cornec D, Chizzolini C, Pers JO, Makowska Z, Lesche R, Kerick M, Alarcón-Riquelme ME, and Martin J
- Subjects
- Adult, Aged, Cohort Studies, Europe, Female, Gene Expression Profiling, Genome-Wide Association Study, Humans, Immunophenotyping, Interferon Type I blood, Male, Microarray Analysis, Middle Aged, Sequence Analysis, RNA, Toll-Like Receptors blood, Autoimmunity genetics, Scleroderma, Systemic genetics, Signal Transduction genetics, Transcriptome genetics
- Abstract
Objectives: The analysis of annotated transcripts from genome-wide expression studies may help to understand the pathogenesis of complex diseases, such as systemic sclerosis (SSc). We performed a whole blood (WB) transcriptome analysis on RNA collected in the context of the European PRECISESADS project, aiming at characterising the pathways that differentiate SSc from controls and that are reproducible in geographically diverse populations., Methods: Samples from 162 patients and 252 controls were collected in RNA stabilisers. Cases and controls were divided into a discovery (n=79+163; Southern Europe) and validation cohort (n=83+89; Central-Western Europe). RNA sequencing was performed by an Illumina assay. Functional annotations of Reactome pathways were performed with the Functional Analysis of Individual Microarray Expression (FAIME) algorithm. In parallel, immunophenotyping of 28 circulating cell populations was performed. We tested the presence of differentially expressed genes/pathways and the correlation between absolute cell counts and RNA transcripts/FAIME scores in regression models. Results significant in both populations were considered as replicated., Results: Overall, 15 224 genes and 1277 functional pathways were available; of these, 99 and 225 were significant in both sets. Among replicated pathways, we found a deregulation in type-I interferon, Toll-like receptor cascade, tumour suppressor p53 protein function, platelet degranulation and activation. RNA transcripts or FAIME scores were jointly correlated with cell subtypes with strong geographical differences; neutrophils were the major determinant of gene expression in SSc-WB samples., Conclusions: We discovered a set of differentially expressed genes/pathways validated in two independent sets of patients with SSc, highlighting a number of deregulated processes that have relevance for the pathogenesis of autoimmunity and SSc., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2020
- Full Text
- View/download PDF
42. Epigenome-Wide Comparative Study Reveals Key Differences Between Mixed Connective Tissue Disease and Related Systemic Autoimmune Diseases.
- Author
-
Carnero-Montoro E, Barturen G, Povedano E, Kerick M, Martinez-Bueno M, Ballestar E, Martin J, Teruel M, and Alarcón-Riquelme ME
- Subjects
- Adult, Autoimmune Diseases genetics, Autoimmune Diseases immunology, DNA Methylation immunology, Female, Genetic Markers immunology, Genome-Wide Association Study, Humans, Male, Middle Aged, DNA Methylation genetics, Genetic Markers genetics, Mixed Connective Tissue Disease genetics, Mixed Connective Tissue Disease immunology
- Abstract
Mixed Connective Tissue Disease (MCTD) is a rare complex systemic autoimmune disease (SAD) characterized by the presence of increased levels of anti-U1 ribonucleoprotein autoantibodies and signs and symptoms that resemble other SADs such as systemic sclerosis (SSc), rheumatoid arthritis (RA), and systemic lupus erythematosus (SLE). Due to its low prevalence, this disease has been very poorly studied at the molecular level. We performed for the first time an epigenome-wide association study interrogating DNA methylation data obtained with the Infinium MethylationEPIC array from whole blood samples in 31 patients diagnosed with MCTD and 255 healthy subjects. We observed a pervasive hypomethylation involving 170 genes enriched for immune-related function such as those involved in type I interferon signaling pathways or in negative regulation of viral genome replication. We mostly identified epigenetic signals at genes previously implicated in other SADs, for example MX1, PARP9, DDX60 , or IFI44L , for which we also observed that MCTD patients exhibit higher DNA methylation variability compared with controls, suggesting that these sites might be involved in plastic immune responses that are relevant to the disease. Through methylation quantitative trait locus (meQTL) analysis we identified widespread local genetic effects influencing DNA methylation variability at MCTD-associated sites. Interestingly, for IRF7, IFI44 genes, and the HLA region we have evidence that they could be exerting a genetic risk on MCTD mediated through DNA methylation changes. Comparison of MCTD-associated epigenome with patients diagnosed with SLE, or Sjögren's Syndrome, reveals a common interferon-related epigenetic signature, however we find substantial epigenetic differences when compared with patients diagnosed with rheumatoid arthritis and systemic sclerosis. Furthermore, we show that MCTD-associated CpGs are potential epigenetic biomarkers with high diagnostic value. Our study serves to reveal new genes and pathways involved in MCTD, to illustrate the important role of epigenetic modifications in MCTD pathology, in mediating the interaction between different genetic and environmental MCTD risk factors, and as potential biomarkers of SADs.
- Published
- 2019
- Full Text
- View/download PDF
43. Genome-wide meta-analysis reveals shared new loci in systemic seropositive rheumatic diseases.
- Author
-
Acosta-Herrera M, Kerick M, González-Serna D, Wijmenga C, Franke A, Gregersen PK, Padyukov L, Worthington J, Vyse TJ, Alarcón-Riquelme ME, Mayes MD, and Martin J
- Subjects
- Adult, Arthritis, Rheumatoid immunology, Female, Genetic Predisposition to Disease, Genome-Wide Association Study, Humans, Lim Kinases immunology, Lupus Erythematosus, Systemic immunology, Male, Membrane Proteins immunology, Myositis immunology, Polymorphism, Single Nucleotide, Quantitative Trait Loci immunology, Repressor Proteins immunology, Rheumatic Diseases immunology, Scleroderma, Systemic immunology, White People genetics, alpha Karyopherins immunology, Arthritis, Rheumatoid genetics, Lupus Erythematosus, Systemic genetics, Myositis genetics, Quantitative Trait Loci genetics, Rheumatic Diseases genetics, Scleroderma, Systemic genetics
- Abstract
Objective: Immune-mediated inflammatory diseases (IMIDs) are heterogeneous and complex conditions with overlapping clinical symptoms and elevated familial aggregation, which suggests the existence of a shared genetic component. In order to identify this genetic background in a systematic fashion, we performed the first cross-disease genome-wide meta-analysis in systemic seropositive rheumatic diseases, namely, systemic sclerosis, systemic lupus erythematosus, rheumatoid arthritis and idiopathic inflammatory myopathies., Methods: We meta-analysed ~6.5 million single nucleotide polymorphisms in 11 678 cases and 19 704 non-affected controls of European descent populations. The functional roles of the associated variants were interrogated using publicly available databases., Results: Our analysis revealed five shared genome-wide significant independent loci that had not been previously associated with these diseases: NAB1 , KPNA4-ARL14 , DGQK , LIMK1 and PRR12 . All of these loci are related with immune processes such as interferon and epidermal growth factor signalling, response to methotrexate, cytoskeleton dynamics and coagulation cascade. Remarkably, several of the associated loci are known key players in autoimmunity, which supports the validity of our results. All the associated variants showed significant functional enrichment in DNase hypersensitivity sites, chromatin states and histone marks in relevant immune cells, including shared expression quantitative trait loci . Additionally, our results were significantly enriched in drugs that are being tested for the treatment of the diseases under study., Conclusions: We have identified shared new risk loci with functional value across diseases and pinpoint new potential candidate loci that could be further investigated. Our results highlight the potential of drug repositioning among related systemic seropositive rheumatic IMIDs., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2019. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2019
- Full Text
- View/download PDF
44. Meta-analysis of Immunochip data of four autoimmune diseases reveals novel single-disease and cross-phenotype associations.
- Author
-
Márquez A, Kerick M, Zhernakova A, Gutierrez-Achury J, Chen WM, Onengut-Gumuscu S, González-Álvaro I, Rodriguez-Rodriguez L, Rios-Fernández R, González-Gay MA, Mayes MD, Raychaudhuri S, Rich SS, Wijmenga C, and Martín J
- Subjects
- Humans, Arthritis, Rheumatoid genetics, Celiac Disease genetics, Diabetes Mellitus, Type 1 genetics, Genome-Wide Association Study, Molecular Sequence Annotation, Scleroderma, Systemic genetics, Autoimmune Diseases genetics, Genetic Predisposition to Disease, Polymorphism, Single Nucleotide
- Abstract
Background: In recent years, research has consistently proven the occurrence of genetic overlap across autoimmune diseases, which supports the existence of common pathogenic mechanisms in autoimmunity. The objective of this study was to further investigate this shared genetic component., Methods: For this purpose, we performed a cross-disease meta-analysis of Immunochip data from 37,159 patients diagnosed with a seropositive autoimmune disease (11,489 celiac disease (CeD), 15,523 rheumatoid arthritis (RA), 3477 systemic sclerosis (SSc), and 6670 type 1 diabetes (T1D)) and 22,308 healthy controls of European origin using the R package ASSET., Results: We identified 38 risk variants shared by at least two of the conditions analyzed, five of which represent new pleiotropic loci in autoimmunity. We also identified six novel genome-wide associations for the diseases studied. Cell-specific functional annotations and biological pathway enrichment analyses suggested that pleiotropic variants may act by deregulating gene expression in different subsets of T cells, especially Th17 and regulatory T cells. Finally, drug repositioning analysis evidenced several drugs that could represent promising candidates for CeD, RA, SSc, and T1D treatment., Conclusions: In this study, we have been able to advance in the knowledge of the genetic overlap existing in autoimmunity, thus shedding light on common molecular mechanisms of disease and suggesting novel drug targets that could be explored for the treatment of the autoimmune diseases studied.
- Published
- 2018
- Full Text
- View/download PDF
45. Genomic loss of HLA alleles may affect the clinical outcome in low-risk myelodysplastic syndrome patients.
- Author
-
Montes P, Kerick M, Bernal M, Hernández F, Jiménez P, Garrido P, Márquez A, Jurado M, Martin J, Garrido F, and Ruiz-Cabello F
- Abstract
The Revised International Prognostic Score and some somatic mutations in myelodysplastic syndrome (MDS) are independently associated with transformation to acute myeloid leukemia (AML). Immunity has also been implicated in the pathogenesis of MDS, although the underlying mechanism remains unclear. We performed a SNP array on chromosome 6 in CD34
+ purified blasts from 19 patients diagnosed with advanced MDS and 8 patients with other myeloid malignancies to evaluate the presence of loss of heterozygosity (LOH) in HLA and its impact on disease progression. Three patients had acquired copy-neutral LOH (CN-LOH) on 6p arms, which may disrupt antigen presentation and act as a mechanism for immune system evasion. Interestingly, these patients had previously been classified at low risk of AML progression, and the poor outcome cannot be explained by the acquisition of adverse mutations. LOH HLA was not detected in the remaining 24 patients, who all had adverse risk factors. In summary, the clinical outcome of patients with advanced MDS might be influenced by HLA allelic loss, wich allows subclonal expansions to evade cytotoxic-T and NK cell attack. CN-LOH HLA may therefore be a factor favoring MDS progression to AML independently of the somatic tumor mutation load., Competing Interests: CONFLICTS OF INTEREST No potential conflicts of interest were disclosed.- Published
- 2018
- Full Text
- View/download PDF
46. Author Correction: LSD1 modulates the non-canonical integrin β3 signaling pathway in non-small cell lung carcinoma cells.
- Author
-
Lim SY, Macheleidt I, Dalvi P, Schäfer SC, Kerick M, Ozretić L, Ortiz-Cuaran S, George J, Merkelbach-Bruse S, Wolf J, Timmermann B, Thomas RK, Schweiger MR, Buettner R, and Odenthal M
- Abstract
A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.
- Published
- 2018
- Full Text
- View/download PDF
47. Epigenomic profiling of non-small cell lung cancer xenografts uncover LRP12 DNA methylation as predictive biomarker for carboplatin resistance.
- Author
-
Grasse S, Lienhard M, Frese S, Kerick M, Steinbach A, Grimm C, Hussong M, Rolff J, Becker M, Dreher F, Schirmer U, Boerno S, Ramisch A, Leschber G, Timmermann B, Grohé C, Lüders H, Vingron M, Fichtner I, Klein S, Odenthal M, Büttner R, Lehrach H, Sültmann H, Herwig R, and Schweiger MR
- Subjects
- Animals, Biomarkers, Tumor metabolism, Carboplatin pharmacology, Disease-Free Survival, Drug Resistance, Neoplasm genetics, Genes, Tumor Suppressor, Genome, Human, Humans, Low Density Lipoprotein Receptor-Related Protein-1 metabolism, Lung Neoplasms genetics, Mice, Nude, Promoter Regions, Genetic, Treatment Outcome, Biomarkers, Tumor genetics, Carboplatin therapeutic use, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung genetics, DNA Methylation genetics, Epigenomics, Low Density Lipoprotein Receptor-Related Protein-1 genetics, Xenograft Model Antitumor Assays
- Abstract
Background: Non-small cell lung cancer (NSCLC) is the most common cause of cancer-related deaths worldwide and is primarily treated with radiation, surgery, and platinum-based drugs like cisplatin and carboplatin. The major challenge in the treatment of NSCLC patients is intrinsic or acquired resistance to chemotherapy. Molecular markers predicting the outcome of the patients are urgently needed., Methods: Here, we employed patient-derived xenografts (PDXs) to detect predictive methylation biomarkers for platin-based therapies. We used MeDIP-Seq to generate genome-wide DNA methylation profiles of 22 PDXs, their parental primary NSCLC, and their corresponding normal tissues and complemented the data with gene expression analyses of the same tissues. Candidate biomarkers were validated with quantitative methylation-specific PCRs (qMSP) in an independent cohort., Results: Comprehensive analyses revealed that differential methylation patterns are highly similar, enriched in PDXs and lung tumor-specific when comparing differences in methylation between PDXs versus primary NSCLC. We identified a set of 40 candidate regions with methylation correlated to carboplatin response and corresponding inverse gene expression pattern even before therapy. This analysis led to the identification of a promoter CpG island methylation of LDL receptor-related protein 12 (LRP12) associated with increased resistance to carboplatin. Validation in an independent patient cohort (n = 35) confirmed that LRP12 methylation status is predictive for therapeutic response of NSCLC patients to platin therapy with a sensitivity of 80% and a specificity of 84% (p < 0.01). Similarly, we find a shorter survival time for patients with LRP12 hypermethylation in the TCGA data set for NSCLC (lung adenocarcinoma)., Conclusions: Using an epigenome-wide sequencing approach, we find differential methylation patterns from primary lung cancer and PDX-derived cancers to be very similar, albeit with a lower degree of differential methylation in primary tumors. We identify LRP12 DNA methylation as a powerful predictive marker for carboplatin resistance. These findings outline a platform for the identification of epigenetic therapy resistance biomarkers based on PDX NSCLC models.
- Published
- 2018
- Full Text
- View/download PDF
48. HLA class I loss and PD-L1 expression in lung cancer: impact on T-cell infiltration and immune escape.
- Author
-
Perea F, Sánchez-Palencia A, Gómez-Morales M, Bernal M, Concha Á, García MM, González-Ramírez AR, Kerick M, Martin J, Garrido F, Ruiz-Cabello F, and Aptsiauri N
- Abstract
Immune-checkpoint inhibitors show encouraging results in cancer treatment, but the clinical benefit is limited exclusively to a subset of patients. We analyzed the density and composition of tumor T-cell infiltration in non-small-cell lung carcinoma (NSCLC) in relation to PD-L1 and HLA class I (HLA-I) expression. We found that positive HLA-I expression, independently on PD-L1 status, is the key factor determining the increased density of the immune infiltrate. When both markers were analyzed simultaneously, we identified four phenotypes of HLA-I and PD-L1 co-expression. They demonstrated different patterns of tumor infiltration and clinicopathologic characteristics, including the tumor size and lymphatic spread. All HLA-I+/PD-L1+ tumors had a high degree of intratumoral infiltration with CD8+T-lymphocytes, whereas HLA-I loss was associated with a significantly reduced number of tumor infiltrating T-lymphocytes mostly restrained in the stroma surrounding the tumor nest. HLA-I-negative/PD-L1-positive tumors had bigger size (T) and lower grade of infiltration with CD8+T-cells. It represents a cancer immune escape phenotype that combines two independent mechanisms of immune evasion: loss of HLA-I and upregulation of PD-L1. Using GCH-array analysis of human lung cancer cell lines we found that the loss of heterozygosity (LOH) with complete or partial deletion of HLA-I genes is the principal mechanism of HLA-I alterations. This irreversible defect, which could potentially decrease the clinical efficacy of lung cancer immunotherapy, appears to be underestimated. In conclusion, our results suggest that the analysis of HLA-I is very important for the selection of potential responders to cancer immunotherapy., Competing Interests: CONFLICTS OF INTEREST The authors have no conflicts of interest to disclose.
- Published
- 2017
- Full Text
- View/download PDF
49. Epithelial barrier dysfunction in lymphocytic colitis through cytokine-dependent internalization of claudin-5 and -8.
- Author
-
Barmeyer C, Erko I, Awad K, Fromm A, Bojarski C, Meissner S, Loddenkemper C, Kerick M, Siegmund B, Fromm M, Schweiger MR, and Schulzke JD
- Subjects
- Adult, Aged, Apoptosis physiology, Blotting, Western, Case-Control Studies, Claudin-4 metabolism, Cytokines metabolism, Diarrhea etiology, Dielectric Spectroscopy methods, Down-Regulation, Female, HT29 Cells, Humans, Male, Microscopy, Confocal, Middle Aged, Sodium metabolism, Tight Junctions metabolism, Claudin-5 metabolism, Claudins metabolism, Colitis, Lymphocytic physiopathology, Intestinal Mucosa pathology
- Abstract
Background: Watery diarrhea is the cardinal symptom of lymphocytic colitis (LC). We have previously shown that colonic Na malabsorption is one of the major pathologic alterations of LC and found evidence for an epithelial barrier defect. On these grounds, this study aimed to identify the inherent mechanisms of this epithelial barrier dysfunction and its regulatory features., Methods: Epithelial resistance (R
epi ) was determined by one-path impedance spectroscopy and3 H-mannitol fluxes were performed on biopsies from sigmoid colon in miniaturized Ussing chambers. Tight junction proteins were analyzed by Western blot and confocal microscopy. Inflammatory signaling was characterized in HT-29/B6 cells. Apoptosis and mucosal surface parameters were quantified morphologically., Results: Repi was reduced to 53% and3 H-mannitol fluxes increased 1.7-fold in LC due to lower expression of claudin-4, -5, and -8 and altered subcellular claudin-5 and -8 distributions off the tight junction. TNFα and IFNγ could mimic subcellular redistribution in HT-29/B6 cells, a process which was independent on MLCK activation. Epithelial apoptosis did not contribute to barrier dysfunction in LC and mucosal surface area was unchanged., Conclusions: Epithelial barrier dysfunction in LC occurs through downregulation of claudin-4, -5, and -8, and redistribution of claudin-5 and -8 off the tight junction, which contributes to diarrhea by a leak-flux mechanism. The key effector cytokines TNFα and IFNγ turned out to be the trigger for redistribution of claudin-5 and -8. Thus, alongside sodium malabsorption, leak-flux is yet another important diarrheal mechanism in LC.- Published
- 2017
- Full Text
- View/download PDF
50. Genomic and transcriptomic heterogeneity of colorectal tumours arising in Lynch syndrome.
- Author
-
Binder H, Hopp L, Schweiger MR, Hoffmann S, Jühling F, Kerick M, Timmermann B, Siebert S, Grimm C, Nersisyan L, Arakelyan A, Herberg M, Buske P, Loeffler-Wirth H, Rosolowski M, Engel C, Przybilla J, Peifer M, Friedrichs N, Moeslein G, Odenthal M, Hussong M, Peters S, Holzapfel S, Nattermann J, Hueneburg R, Schmiegel W, Royer-Pokora B, Aretz S, Kloth M, Kloor M, Buettner R, Galle J, and Loeffler M
- Subjects
- Antigens, Neoplasm genetics, Colorectal Neoplasms immunology, Colorectal Neoplasms, Hereditary Nonpolyposis genetics, Colorectal Neoplasms, Hereditary Nonpolyposis immunology, Gene Expression genetics, Genes, Neoplasm genetics, Genome, Human genetics, Humans, Immunity, Cellular, Phenotype, Recurrence, Transcriptome genetics, Tumor Escape genetics, Tumor Escape immunology, Colorectal Neoplasms genetics, Mutation genetics
- Abstract
Colorectal cancer (CRC) arising in Lynch syndrome (LS) comprises tumours with constitutional mutations in DNA mismatch repair genes. There is still a lack of whole-genome and transcriptome studies of LS-CRC to address questions about similarities and differences in mutation and gene expression characteristics between LS-CRC and sporadic CRC, about the molecular heterogeneity of LS-CRC, and about specific mechanisms of LS-CRC genesis linked to dysfunctional mismatch repair in LS colonic mucosa and the possible role of immune editing. Here, we provide a first molecular characterization of LS tumours and of matched tumour-distant reference colonic mucosa based on whole-genome DNA-sequencing and RNA-sequencing analyses. Our data support two subgroups of LS-CRCs, G1 and G2, whereby G1 tumours show a higher number of somatic mutations, a higher amount of microsatellite slippage, and a different mutation spectrum. The gene expression phenotypes support this difference. Reference mucosa of G1 shows a strong immune response associated with the expression of HLA and immune checkpoint genes and the invasion of CD4+ T cells. Such an immune response is not observed in LS tumours, G2 reference and normal (non-Lynch) mucosa, and sporadic CRC. We hypothesize that G1 tumours are edited for escape from a highly immunogenic microenvironment via loss of HLA presentation and T-cell exhaustion. In contrast, G2 tumours seem to develop in a less immunogenic microenvironment where tumour-promoting inflammation parallels tumourigenesis. Larger studies on non-neoplastic mucosa tissue of mutation carriers are required to better understand the early phases of emerging tumours. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd., (Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.)
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.