1
|
Azuaga AB, Ramírez J, Cañete JD. Psoriatic Arthritis: Pathogenesis and Targeted Therapies. Int J Mol Sci 2023; 24:4901. [PMID: 36902329 PMCID: PMC10003101 DOI: 10.3390/ijms24054901] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
Psoriatic arthritis (PsA), a heterogeneous chronic inflammatory immune-mediated disease characterized by musculoskeletal inflammation (arthritis, enthesitis, spondylitis, and dactylitis), generally occurs in patients with psoriasis. PsA is also associated with uveitis and inflammatory bowel disease (Crohn's disease and ulcerative colitis). To capture these manifestations as well as the associated comorbidities, and to recognize their underlining common pathogenesis, the name of psoriatic disease was coined. The pathogenesis of PsA is complex and multifaceted, with an interplay of genetic predisposition, triggering environmental factors, and activation of the innate and adaptive immune system, although autoinflammation has also been implicated. Research has identified several immune-inflammatory pathways defined by cytokines (IL-23/IL-17, TNF), leading to the development of efficacious therapeutic targets. However, heterogeneous responses to these drugs occur in different patients and in the different tissues involved, resulting in a challenge to the global management of the disease. Therefore, more translational research is necessary in order to identify new targets and improve current disease outcomes. Hopefully, this may become a reality through the integration of different omics technologies that allow better understanding of the relevant cellular and molecular players of the different tissues and manifestations of the disease. In this narrative review, we aim to provide an updated overview of the pathophysiology, including the latest findings from multiomics studies, and to describe current targeted therapies.
Collapse
Affiliation(s)
- Ana Belén Azuaga
- Rheumatology Department, Hospital Clinic and IDIBAPS of Barcelona, 08036 Barcelona, Spain
| | | | - Juan D. Cañete
- Rheumatology Department, Hospital Clinic and IDIBAPS of Barcelona, 08036 Barcelona, Spain
| |
Collapse
|
2
|
Splice factor polypyrimidine tract-binding protein 1 (Ptbp1) primes endothelial inflammation in atherogenic disturbed flow conditions. Proc Natl Acad Sci U S A 2022; 119:e2122227119. [PMID: 35858420 PMCID: PMC9335344 DOI: 10.1073/pnas.2122227119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Plaque forms in low and disturbed flow regions of the vasculature, where platelets adhere and endothelial cells are “primed” to respond to cytokines (e.g., tumor necrosis factor-α) with elevated levels of cell adhesion molecules via the NF-κB signaling pathway. We show that the splice factor polypyrimidine tract binding protein (Ptbp1; purple) mediates priming. Ptbp1 is induced in endothelial cells by platelet recruitment, promoting priming and subsequent myeloid cell infiltration into plaque. Mechanistically, Ptbp1 regulates splicing of genes (e.g., Ripk1) involved in the NF-κB signaling pathway and is required for efficient nuclear translocation of NF-κB in endothelial cells. This provides new insight into the molecular mechanisms underlying an endothelial priming process that reinforces vascular inflammation. NF-κB–mediated endothelial activation drives leukocyte recruitment and atherosclerosis, in part through adhesion molecules Icam1 and Vcam1. The endothelium is primed for cytokine activation of NF-κB by exposure to low and disturbed blood flow (LDF)but the molecular underpinnings are not fully understood. In an experimental in vivo model of LDF, platelets were required for the increased expression of several RNA-binding splice factors, including polypyrimidine tract binding protein (Ptbp1). This was coordinated with changes in RNA splicing in the NF-κB pathway in primed cells, leading us to examine splice factors as mediators of priming. Using Icam1 and Vcam1 induction by tumor necrosis factor (TNF)-α stimulation as a readout, we performed a CRISPR Cas9 knockout screen and identified a requirement for Ptbp1 in priming. Deletion of Ptbp1 had no effect on cell growth or response to apoptotic stimuli, but reversed LDF splicing patterns and inhibited NF-κB nuclear translocation and transcriptional activation of downstream targets, including Icam1 and Vcam1. In human coronary arteries, elevated PTBP1 correlates with expression of TNF pathway genes and plaque. In vivo, endothelial-specific deletion of Ptbp1 reduced Icam1 expression and myeloid cell infiltration at regions of LDF in atherosclerotic mice, limiting atherosclerosis. This may be mediated, in part, by allowing inclusion of a conserved alternative exon in Ripk1 leading to a reduction in Ripk1 protein. Our data show that Ptbp1, which is induced in a subset of the endothelium by platelet recruitment at regions of LDF, is required for priming of the endothelium for subsequent NF-κB activation, myeloid cell recruitment and atherosclerosis.
Collapse
|
3
|
Grivas A, Fragoulis G, Garantziotis P, Banos A, Nikiphorou E, Boumpas D. Unraveling the complexities of psoriatic arthritis by the use of -Omics and their relevance for clinical care. Autoimmun Rev 2021; 20:102949. [PMID: 34509654 DOI: 10.1016/j.autrev.2021.102949] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 06/30/2021] [Indexed: 12/30/2022]
Abstract
-Omic technologies represent a novel approach to unravel ill-defined aspects of psoriatic arthritis (PsA). Large-scale information can be acquired from analysis of affected tissues in PsA via high-throughput studies in the domains of genomics, transcriptomics, epigenetics, proteomics and metabolomics. This is a critical overview of the current knowledge of -omics in PsA, with emphasis on the pathophysiological insights of diagnostic and therapeutic relevance, the advent of novel biomarkers and their potential use for precision medicine in PsA.
Collapse
Affiliation(s)
- Alexandros Grivas
- National and Kapodistrian University of Athens, Faculty of medicine, Athens, Greece; Inflammation & Autoimmunity Lab, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece.
| | - George Fragoulis
- First Department of Propaedeutic Internal Medicine, National and Kapodistrian University of Athens, "Laiko" General Hospital, Athens, Greece
| | - Panagiotis Garantziotis
- Inflammation & Autoimmunity Lab, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece; Division of Immunology and Rheumatology, Hannover Medical University, 30,625 Hannover, Germany
| | - Aggelos Banos
- Inflammation & Autoimmunity Lab, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - Elena Nikiphorou
- Centre for Rheumatic Diseases, School of Immunology and Microbial Sciences, King's College London, King's Hospital, London, United Kingdom
| | - Dimitrios Boumpas
- National and Kapodistrian University of Athens, Faculty of medicine, Athens, Greece; Inflammation & Autoimmunity Lab, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| |
Collapse
|
4
|
Zhen Q, Zhang Y, Yu Y, Yang H, Zhang T, Li X, Mo X, Li B, Wu J, Liang Y, Ge H, Xu Q, Chen W, Qian W, Xu H, Chen G, Bai B, Zhang J, Lu Y, Chen S, Zhang H, Zhang Y, Chen X, Li X, Jin X, Lin X, Yong L, Fang M, Zhao J, Lu Y, Wu S, Jiang D, Shi J, Cao H, Qiu Y, Li S, Kang X, Shen J, Ma H, Sun S, Fan Y, Chen W, Bai M, Jiang Q, Li W, Lv C, Li S, Chen M, Li F, Li Y, Sun L. Three Novel Structural Variations at MHC and IL12B Predisposing to Psoriasis. Br J Dermatol 2021; 186:307-317. [PMID: 34498260 DOI: 10.1111/bjd.20752] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Structural variations (SVs, defined as DNA variants ≥50 bp) have been associated with various complex human diseases. However, research to screen the whole genome for SVs predisposing to psoriasis is still lacking. OBJECTIVES This study aimed to investigate the association of SVs and psoriasis. METHODS We performed a genome-wide screen on SVs using an imputation method on 5 independent cohorts with 45,386 subjects from the Chinese Han population. Fine mapping analysis, genetic interaction analysis and RNA expression analysis were conducted to explore the mechanism of SVs. RESULTS We obtained 4,535 SVs in total and identified 2 novel deletions (esv3608550, OR=2.73, P<2.00×10-308 ; esv3608542, OR=0.47, P=7.40×10-28 ) at 6q21.33 (MHC), 1 novel Alu element insertion (esv3607339, OR=1.22, P=1.18×10-35 ) at 5q33.3 (IL12B), and confirmed 1 previously reported deletion (esv3587563, OR=1.30, P=9.52×10-60 ) at 1q21.2 (LCE) for psoriasis. Fine mapping analysis including SNPs and small Insertions/Deletions (InDels) revealed that esv3608550 and esv3608542 were independently associated with psoriasis, and a novel independent SNP (rs9378188, OR=1.65, P=3.46×10-38 ) was identified at 6q21.33. By genetic interaction analysis and RNA expression analysis, we speculate that the association of 2 deletions at 6q21.33 with psoriasis might relate to their influence on the expression of HLA-C. CONCLUSIONS Our study constructed the most comprehensive SV map for psoriasis thus far and enriched the genetic architecture and pathogenesis of psoriasis as well as highlighted the nonnegligible impact of SVs on complex diseases.
Collapse
Affiliation(s)
- Q Zhen
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China, 230032.,Anhui Provincial Institute of Translational Medicine, Hefei, 230032, China
| | - Y Zhang
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Y Yu
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China, 230032.,Anhui Provincial Institute of Translational Medicine, Hefei, 230032, China
| | - H Yang
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - T Zhang
- Department of Biology, University of Copenhagen, Ole MaalØes Vej 5, 2200, Copenhagen, Denmark
| | - X Li
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - X Mo
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - B Li
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China.,The Comprehensive Lab, College of Basic, Anhui Medical University
| | - J Wu
- Department of Dermatology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University
| | - Y Liang
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - H Ge
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China, 230032.,Anhui Provincial Institute of Translational Medicine, Hefei, 230032, China
| | - Q Xu
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China, 230032.,Anhui Provincial Institute of Translational Medicine, Hefei, 230032, China
| | - W Chen
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China, 230032.,Anhui Provincial Institute of Translational Medicine, Hefei, 230032, China
| | - W Qian
- Institute of Dermalology, Guangzhou Medical University, Guangzhou, 510095, China
| | - H Xu
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - G Chen
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China, 230032.,Anhui Provincial Institute of Translational Medicine, Hefei, 230032, China
| | - B Bai
- Department of Dermatology at No.2 Hospital, Harbin Medical University, Harbin, Heilongjiang, 150001, China
| | - J Zhang
- Department of Dermatology, The 195 Hospital of Chinese People's Liberation Army, Xianning, Hubei, 437100, China
| | - Y Lu
- Dermatology Department of the First Affiliated Hospital, Nanjng Medical University, Nanjing, Jiangsu, 210029, China
| | - S Chen
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China, 230032.,Anhui Provincial Institute of Translational Medicine, Hefei, 230032, China
| | - H Zhang
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China, 230032.,Anhui Provincial Institute of Translational Medicine, Hefei, 230032, China
| | - Y Zhang
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - X Chen
- Department of Dermatology at Chengdu Second People's Hospital, Sichuan, Chengdu, 610017, China
| | - X Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - X Jin
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China
| | - X Lin
- Department of Neurology and Institute of Neurology, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China
| | - L Yong
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China, 230032.,Anhui Provincial Institute of Translational Medicine, Hefei, 230032, China
| | - M Fang
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture, Fisheries College, Jimei University, Xiamen, Fujian, 361021, China
| | - J Zhao
- Department of Dermatology, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, Urumqi, 830001, China
| | - Y Lu
- Department of Dermatology at Chengdu Second People's Hospital, Sichuan, Chengdu, 610017, China
| | - S Wu
- Urology Institute of Shenzhen University, The Luohu Affiliated Hospital of Shenzhen University
| | - D Jiang
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture, Fisheries College, Jimei University, Xiamen, Fujian, 361021, China
| | - J Shi
- Department of Dermatology at the Second Affiliated Hospital, Baotou Medical College, University Of Science and Technology Of The Inner Mongolia, Baotou, Inner Mongolia, 014030, China
| | - H Cao
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Y Qiu
- Department of Dermatology, Jining No. 1 People's Hospital, Shandong, 272011, China
| | - S Li
- Department of Dermatology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - X Kang
- Department of Dermatology, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, Urumqi, 830001, China
| | - J Shen
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - H Ma
- Department of Dematology, the 2rd Hospital of Xi'an Jiaotong University. Xi'an, Shanxi, 710004, China
| | - S Sun
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Y Fan
- Department of Dermatology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - W Chen
- Department of Neurology and Institute of Neurology, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China
| | - M Bai
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Q Jiang
- Donggang Center Hospital, Dandong, Liaoning, 118300
| | - W Li
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, Shandong, 272067, China
| | - C Lv
- Dalian Dermatosis Hospital, Dalian, Liaoning, 116021, China
| | - S Li
- Department of Dermatology at No, Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - M Chen
- Dermatology Hospital, Peking Union Medical College
| | - F Li
- Department of Dermatology, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Y Li
- Department of Dermatology, The 195 Hospital of Chinese People's Liberation Army, Xianning, Hubei, 437100, China
| | - L Sun
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China, 230032.,Anhui Provincial Institute of Translational Medicine, Hefei, 230032, China
| |
Collapse
|
5
|
MAGI1, a Scaffold Protein with Tumor Suppressive and Vascular Functions. Cells 2021; 10:cells10061494. [PMID: 34198584 PMCID: PMC8231924 DOI: 10.3390/cells10061494] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 12/13/2022] Open
Abstract
MAGI1 is a cytoplasmic scaffolding protein initially identified as a component of cell-to-cell contacts stabilizing cadherin-mediated cell–cell adhesion in epithelial and endothelial cells. Clinical-pathological and experimental evidence indicates that MAGI1 expression is decreased in some inflammatory diseases, and also in several cancers, including hepatocellular carcinoma, colorectal, cervical, breast, brain, and gastric cancers and appears to act as a tumor suppressor, modulating the activity of oncogenic pathways such as the PI3K/AKT and the Wnt/β-catenin pathways. Genomic mutations and other mechanisms such as mechanical stress or inflammation have been described to regulate MAGI1 expression. Intriguingly, in breast and colorectal cancers, MAGI1 expression is induced by non-steroidal anti-inflammatory drugs (NSAIDs), suggesting a role in mediating the tumor suppressive activity of NSAIDs. More recently, MAGI1 was found to localize at mature focal adhesion and to regulate integrin-mediated adhesion and signaling in endothelial cells. Here, we review MAGI1′s role as scaffolding protein, recent developments in the understanding of MAGI1 function as tumor suppressor gene, its role in endothelial cells and its implication in cancer and vascular biology. We also discuss outstanding questions about its regulation and potential translational implications in oncology.
Collapse
|
6
|
Mulder MLM, van Hal TW, Wenink MH, Koenen HJPM, van den Hoogen FHJ, de Jong EMGJ, van den Reek JMPA, Vriezekolk JE. Clinical, laboratory, and genetic markers for the development or presence of psoriatic arthritis in psoriasis patients: a systematic review. Arthritis Res Ther 2021; 23:168. [PMID: 34127053 PMCID: PMC8201808 DOI: 10.1186/s13075-021-02545-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 05/21/2021] [Indexed: 12/11/2022] Open
Abstract
Twenty to thirty percent of psoriasis (Pso) patients will develop psoriatic arthritis (PsA). Detection of Pso patients that are (at risk for) developing PsA is essential to prevent structural damage. We conducted a systematic search of five bibliographic databases, up to May 2020. We searched for studies assessing markers (clinical, laboratory, genetic) associated with the development or presence of PsA in Pso patients. Study selection and quality assessment of the included studies was performed, followed by a qualitative best evidence synthesis to determine the level of evidence for a marker and its association with concomitant/developing PsA in Pso. Overall, 259 possible markers were identified in 119 studies that met the inclusion criteria. Laboratory markers related to inflammation and bone metabolism reached a strong level of evidence for the association (not prediction) of PsA in Pso. Only CXCL10 showed strong evidence for a positive predictive value for PsA in Pso. The importance of timely detecting PsA in a Pso population, and finding more (bio)markers contributing to early detection, remains high.
Collapse
Affiliation(s)
- Michelle L M Mulder
- Department of Rheumatology, Sint Maartenskliniek, PO box 9011, 6500 GM, Nijmegen, The Netherlands. .,Radboud Institute for Health Sciences (RIHS), Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Tamara W van Hal
- Department of Rheumatology, Sint Maartenskliniek, PO box 9011, 6500 GM, Nijmegen, The Netherlands.,Radboud Institute for Health Sciences (RIHS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mark H Wenink
- Department of Rheumatology, Sint Maartenskliniek, PO box 9011, 6500 GM, Nijmegen, The Netherlands
| | - Hans J P M Koenen
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Elke M G J de Jong
- Radboud Institute for Health Sciences (RIHS), Radboud University Medical Center, Nijmegen, The Netherlands.,Radboud University, Nijmegen, The Netherlands.,Department of Dermatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Juul M P A van den Reek
- Radboud Institute for Health Sciences (RIHS), Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Dermatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Johanna E Vriezekolk
- Department of Rheumatology, Sint Maartenskliniek, PO box 9011, 6500 GM, Nijmegen, The Netherlands
| |
Collapse
|
7
|
Wu D, Wong P, Lam SHM, Li EK, Qin L, Tam LS, Gu J. The causal effect of interleukin-17 on the risk of psoriatic arthritis: a Mendelian randomization study. Rheumatology (Oxford) 2021; 60:1963-1973. [PMID: 33188428 DOI: 10.1093/rheumatology/keaa629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 07/24/2020] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE To determine causal associations between genetically predicted TNF-α, IL-12p70 and IL-17 levels and risk of PsA. METHODS The publicly available summary-level findings from genome-wide association studies (GWAS) was used to identify loci influencing normal physiological concentrations of TNF-α, IL-12p70 and IL-17 (n = 8293) among healthy individuals as exposure and a GWAS for PsA from the UK Biobank (PsA = 900, control = 462 033) as the outcome. A two-sample Mendelian randomization (MR) analysis was performed using the inverse-variance weighted (IVW), weighted median and MR-Egger regression methods. Sensitivity analysis and MR-Egger regression analysis were performed to evaluate the heterogeneity and pleiotropic effects of each variant. RESULTS Single-nucleotide polymorphisms (SNPs) at genome-wide significance from GWASs on TNF-α, IL-12p70 and IL-17 were identified as the instrumental variables. The IVW method indicated a causal association between increased IL-17 level and risk of PsA (β = -0.00186 per allele, s.e. = 0.00043, P = 0.002). Results were consistent in the weighted median method (β = -0.00145 per allele, s.e. = 0.00059, P = 0.014) although the MR-Egger method suggested a non-significant association (β = -0.00133 per allele, s.e. = 0.00087; P = 0.087). Single SNP MR results revealed that the C allele of rs117556572 was robustly associated with risk of PsA (β = 0.00210, s.e. = 0.00069, P = 0.002). However, no evidence for a causal effect was observed between TNF-α, IL-12p70, decreased IL-17 levels and risk of PsA. CONCLUSION Our findings provide preliminary evidence that genetic variants predisposing to higher physiological IL-17 level are associated with decreased risk of PsA.
Collapse
Affiliation(s)
- Dongze Wu
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Department of Medicine & Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Priscilla Wong
- Department of Medicine & Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Steven H M Lam
- Department of Medicine & Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Edmund K Li
- Department of Medicine & Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Ling Qin
- Bone Quality and Health Centre of the Department of Orthopedics & Traumatology, The Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Lai-Shan Tam
- Department of Medicine & Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Jieruo Gu
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
8
|
Urruticoechea-Arana A, Benavent D, León F, Almodovar R, Belinchón I, de la Cueva P, Fernández-Carballido C, Loza E, Gratacós J. Psoriatic arthritis screening: A systematic literature review and experts' recommendations. PLoS One 2021; 16:e0248571. [PMID: 33720981 PMCID: PMC7959352 DOI: 10.1371/journal.pone.0248571] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 03/01/2021] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVE To analyze the performance of psoriatic arthritis (PsA) screening tools, examine their implementation in daily practice, and reach a consensus about the best screening tool for implementation in daily practice in different medical settings. METHODS A systematic literature review (SLR), structured telephone interviews to hospitals, and a multidisciplinary nominal group meeting were all conducted. The SLR employed sensitive search strategies using Medline, Embase, and the Cochrane Library up to January 2020. Two reviewers independently selected articles that reported data on PsA screening tools and that included sufficient data to at least calculate the sensitivity and specificity of those tools (e.g., questionnaires, algorithms, specific questions, and biomarkers). The hospital interviews collected data regarding the process of suspected PsA diagnosis and referral to rheumatology, the implementation of PsA screening tools, and barriers and facilitators to implementation of those tools. In the nominal group meeting, a multidisciplinary team of experts discussed all these data and subsequently recommended a screening tool for implementation. RESULTS The SLR included 41 moderate-quality studies that analyzed 14 PsA screening tools, most of which were questionnaire-based tools. All of these studies reported a moderate-good performance but presented different characteristics regarding the time to completion or the number and type of items or questions. The implementation of screening tools was low (30.5%). The experts ultimately recommended regular use of a PsA screening tool, preferably the PURE-4 questionnaire. CONCLUSIONS The implementation of PsA screening tools like the PURE-4 questionnaire in daily practice likely improves the prognosis of PsA patients.
Collapse
Affiliation(s)
| | - Diego Benavent
- Rheumatology Department, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain
| | - Fernando León
- Primary Care, San Juan de la Cruz Health Center, Pozuelo de Alarcón, Madrid, Spain
| | - Raquel Almodovar
- Rheumatology Department, Hospital Universitario Fundación Alcorcón, Madrid, Spain
| | - Isabel Belinchón
- Department of Dermatology, Hospital General Universitario de Alicante-ISABIAL-UMH, Alicante, Spain
| | - Pablo de la Cueva
- Dermatology Department, Hospital Universitario Infanta Leonor, Madrid, Spain
| | | | - Estíbaliz Loza
- Instituto de Salud Musculoesquelética, InMusc, Madrid, Spain
| | - Jordi Gratacós
- Rheumatology Department, Hospital Universitari Parc Taulí, Sabadell, Spain
| | | |
Collapse
|
9
|
Insights into the pathogenesis of psoriatic arthritis from genetic studies. Semin Immunopathol 2021; 43:221-234. [PMID: 33712923 DOI: 10.1007/s00281-021-00843-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 01/19/2021] [Indexed: 12/20/2022]
Abstract
Psoriatic arthritis (PsA) is a relatively common inflammatory arthritis, a spondyloarthritis (SpA), that occurs most often in patients with psoriasis, a common immune-mediated inflammatory skin disease. Both psoriasis and PsA are highly heritable. Genetic and recent genomic studies have identified variants associated with psoriasis and PsA, but variants differentiating psoriasis from PsA are few. In this review, we describe recent developments in understanding the genetic burden of PsA, linkage, association and epigenetic studies. Using pathway analysis, we provide further insights into the similarities and differences between PsA and psoriasis, as well as between PsA and other immune-mediated inflammatory diseases, particularly ankylosing spondylitis, another SpA. Environmental factors that may trigger PsA in patients with psoriasis are also reviewed. To further understand the pathogenetic differences between PsA and psoriasis as well as other SpA, larger cohort studies of well-phenotyped subjects with integrated analysis of genomic, epigenomic, transcriptomic, proteomic and metabolomic data using interomic system biology approaches are required.
Collapse
|
10
|
Dehnavi S, Sadeghi M, Penson PE, Banach M, Jamialahmadi T, Sahebkar A. The Role of Protein SUMOylation in the Pathogenesis of Atherosclerosis. J Clin Med 2019; 8:E1856. [PMID: 31684100 PMCID: PMC6912227 DOI: 10.3390/jcm8111856] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 10/27/2019] [Accepted: 10/28/2019] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is a progressive, inflammatory cardiovascular disorder characterized by the development of lipid-filled plaques within arteries. Endothelial cell dysfunction in the walls of blood vessels results in an increase in vascular permeability, alteration of the components of the extracellular matrix, and retention of LDL in the sub-endothelial space, thereby accelerating plaque formation. Epigenetic modification by SUMOylation can influence the surface interactions of target proteins and affect cellular functionality, thereby regulating multiple cellular processes. Small ubiquitin-like modifier (SUMO) can modulate NFκB and other proteins such as p53, KLF, and ERK5, which have critical roles in atherogenesis. Furthermore, SUMO regulates leukocyte recruitment and cytokine release and the expression of adherence molecules. In this review, we discuss the regulation by SUMO and SUMOylation modifications of proteins and pathways involved in atherosclerosis.
Collapse
Affiliation(s)
- Sajad Dehnavi
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 6135715794, Iran.
| | - Mahvash Sadeghi
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran.
| | - Peter E Penson
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool L3 3AF, UK.
| | - Maciej Banach
- Department of Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Zeromskiego 113, Lodz 93-338, Poland.
- Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz 93-338, Poland.
| | - Tannaz Jamialahmadi
- Halal Research Center of IRI, FDA, Tehran, Iran.
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran.
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9188617871, Iran.
| |
Collapse
|
11
|
Abe JI, Ko KA, Kotla S, Wang Y, Paez-Mayorga J, Shin IJ, Imanishi M, Vu HT, Tao Y, Leiva-Juarez MM, Thomas TN, Medina JL, Won JH, Fujii Y, Giancursio CJ, McBeath E, Shin JH, Guzman L, Abe RJ, Taunton J, Mochizuki N, Faubion W, Cooke JP, Fujiwara K, Evans SE, Le NT. MAGI1 as a link between endothelial activation and ER stress drives atherosclerosis. JCI Insight 2019; 4:125570. [PMID: 30944250 DOI: 10.1172/jci.insight.125570] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 02/14/2019] [Indexed: 01/06/2023] Open
Abstract
The possible association between the membrane-associated guanylate kinase with inverted domain structure-1 (MAGI1) and inflammation has been suggested, but the molecular mechanisms underlying this link, especially during atherogenesis, remain unclear. In endothelial cells (ECs) exposed to disturbed flow (d-flow), p90 ribosomal S6 kinase (p90RSK) bound to MAGI1, causing MAGI1-S741 phosphorylation and sentrin/SUMO-specific protease 2 T368 phosphorylation-mediated MAGI1-K931 deSUMOylation. MAGI1-S741 phosphorylation upregulated EC activation via activating Rap1. MAGI1-K931 deSUMOylation induced both nuclear translocation of p90RSK-MAGI1 and ATF-6-MAGI1 complexes, which accelerated EC activation and apoptosis, respectively. Microarray screening revealed key roles for MAGI1 in the endoplasmic reticulum (ER) stress response. In this context, MAGI1 associated with activating transcription factor 6 (ATF-6). MAGI1 expression was upregulated in ECs and macrophages found in atherosclerotic-prone regions of mouse aortas as well as in the colonic epithelia and ECs of patients with inflammatory bowel disease. Further, reduced MAGI1 expression in Magi1-/+ mice inhibited d-flow-induced atherogenesis. In sum, EC activation and ER stress-mediated apoptosis are regulated in concert by two different types of MAGI1 posttranslational modifications, elucidating attractive drug targets for chronic inflammatory disease, particularly atherosclerosis.
Collapse
Affiliation(s)
- Jun-Ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kyung Ae Ko
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sivareddy Kotla
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yin Wang
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jesus Paez-Mayorga
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston Texas, USA.,Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo Leon, Mexico
| | - Ik Jae Shin
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Masaki Imanishi
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hang Thi Vu
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yunting Tao
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston Texas, USA
| | - Miguel M Leiva-Juarez
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Tamlyn N Thomas
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jan L Medina
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jong Hak Won
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yuka Fujii
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Carolyn J Giancursio
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston Texas, USA
| | - Elena McBeath
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ji-Hyun Shin
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Liliana Guzman
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston Texas, USA
| | - Rei J Abe
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston Texas, USA
| | - Jack Taunton
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, California, USA
| | - Naoki Mochizuki
- Department of Cell Biology, National Cardiovascular Center Research Institute, Osaka, Japan
| | - William Faubion
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - John P Cooke
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston Texas, USA
| | - Keigi Fujiwara
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Scott E Evans
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nhat-Tu Le
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston Texas, USA
| |
Collapse
|
12
|
Aterido A, Cañete JD, Tornero J, Ferrándiz C, Pinto JA, Gratacós J, Queiró R, Montilla C, Torre-Alonso JC, Pérez-Venegas JJ, Fernández Nebro A, Muñoz-Fernández S, González CM, Roig D, Zarco P, Erra A, Rodríguez J, Castañeda S, Rubio E, Salvador G, Díaz-Torné C, Blanco R, Willisch Domínguez A, Mosquera JA, Vela P, Sánchez-Fernández SA, Corominas H, Ramírez J, de la Cueva P, Fonseca E, Fernández E, Puig L, Dauden E, Sánchez-Carazo JL, López-Estebaranz JL, Moreno D, Vanaclocha F, Herrera E, Blanco F, Fernández-Gutiérrez B, González A, Pérez-García C, Alperi-López M, Olivé Marques A, Martínez-Taboada V, González-Álvaro I, Sanmartí R, Tomás Roura C, García-Montero AC, Bonàs-Guarch S, Mercader JM, Torrents D, Codó L, Gelpí JL, López-Corbeto M, Pluma A, López-Lasanta M, Tortosa R, Palau N, Absher D, Myers R, Marsal S, Julià A. Genetic variation at the glycosaminoglycan metabolism pathway contributes to the risk of psoriatic arthritis but not psoriasis. Ann Rheum Dis 2018; 78:annrheumdis-2018-214158. [PMID: 30552173 DOI: 10.1136/annrheumdis-2018-214158] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 11/16/2018] [Accepted: 11/16/2018] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Psoriatic arthritis (PsA) is a chronic inflammatory arthritis affecting up to 30% of patients with psoriasis (Ps). To date, most of the known risk loci for PsA are shared with Ps, and identifying disease-specific variation has proven very challenging. The objective of the present study was to identify genetic variation specific for PsA. METHODS We performed a genome-wide association study in a cohort of 835 patients with PsA and 1558 controls from Spain. Genetic association was tested at the single marker level and at the pathway level. Meta-analysis was performed with a case-control cohort of 2847 individuals from North America. To confirm the specificity of the genetic associations with PsA, we tested the associated variation using a purely cutaneous psoriasis cohort (PsC, n=614) and a rheumatoid arthritis cohort (RA, n=1191). Using network and drug-repurposing analyses, we further investigated the potential of the PsA-specific associations to guide the development of new drugs in PsA. RESULTS We identified a new PsA risk single-nucleotide polymorphism at B3GNT2 locus (p=1.10e-08). At the pathway level, we found 14 genetic pathways significantly associated with PsA (pFDR<0.05). From these, the glycosaminoglycan (GAG) metabolism pathway was confirmed to be disease-specific after comparing the PsA cohort with the cohorts of patients with PsC and RA. Finally, we identified candidate drug targets in the GAG metabolism pathway as well as new PsA indications for approved drugs. CONCLUSION These findings provide insights into the biological mechanisms that are specific for PsA and could contribute to develop more effective therapies.
Collapse
Affiliation(s)
- Adrià Aterido
- Rheumatology Research Group, Vall d'Hebron Research Institute, Barcelona, Spain.,Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Juan D Cañete
- Rheumatology Department, Hospital Clínic de Barcelona and IDIBAPS, Barcelona, Spain
| | - Jesús Tornero
- Rheumatology Department, Hospital Universitario Guadalajara, Guadalajara, Spain
| | - Carlos Ferrándiz
- Dermatology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - José Antonio Pinto
- Rheumatology Department, Complejo Hospitalario Juan Canalejo, A Coruña, Spain
| | - Jordi Gratacós
- Rheumatology Department, Hospital Parc Taulí, Sabadell, Spain
| | - Rubén Queiró
- Rheumatology Department, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Carlos Montilla
- Rheumatology Department, Hospital Virgen de la Vega, Salamanca, Spain
| | | | | | - Antonio Fernández Nebro
- Rheumatology Department, Instituto de Investigación Biomédica de Málaga, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Santiago Muñoz-Fernández
- Rheumatology Department, Hospital Universitario Infanta Sofía, Universidad Europea, Madrid, Spain
| | - Carlos M González
- Rheumatology Department, Hospital Universitario Gregorio Marañón, Madrid, Spain
| | - Daniel Roig
- Rheumatology Department, Hospital Moisès Broggi, Barcelona, Spain
| | - Pedro Zarco
- Rheumatology Department, Hospital Universitario Fundación Alcorcón, Madrid, Spain
| | - Alba Erra
- Rheumatology Department, Hospital Sant Rafael, Barcelona, Spain
| | - Jesús Rodríguez
- Rheumatology Department, Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Santos Castañeda
- Rheumatology Department, Hospital Universitario La Princesa, IIS La Princesa, Madrid, Spain
| | - Esteban Rubio
- Rheumatology Department, Centro de Salud Virgen de los Reyes, Sevilla, Spain
| | - Georgina Salvador
- Rheumatology Department, Hospital Universitario Mútua de Terrassa, Terrassa, Spain
| | - Cesar Díaz-Torné
- Rheumatology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Ricardo Blanco
- Rheumatology Department, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | | | - José Antonio Mosquera
- Rheumatology Department, Complejo Hospitalario Hospital Provincial de Pontevedra, Pontevedra, Spain
| | - Paloma Vela
- Rheumatology Department, Hospital General Universitario de Alicante, Alicante, Spain
| | | | - Héctor Corominas
- Rheumatology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Rheumatology Department, Hospital Dos de Maig, Barcelona, Spain
| | - Julio Ramírez
- Rheumatology Department, Hospital Clínic de Barcelona and IDIBAPS, Barcelona, Spain
| | - Pablo de la Cueva
- Dermatology Department, Hospital Universitario Infanta Leonor, Madrid, Spain
| | - Eduardo Fonseca
- Dermatology Department, Complejo Hospitalario Universitario de A Coruña, A Coruña, Spain
| | - Emilia Fernández
- Dermatology Department, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Lluis Puig
- Dermatology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Esteban Dauden
- Dermatology Department, Hospital Universitario La Princesa, IIS La Princesa, Madrid, Spain
| | | | | | - David Moreno
- Dermatology Department, Hospital Virgen Macarena, Sevilla, Spain
| | | | - Enrique Herrera
- Dermatology Department, Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - Francisco Blanco
- Rheumatology Department, INIBIC-Hospital Universitario A Coruña, A Coruña, Spain
| | | | - Antonio González
- Instituto de Investigación Sanitaria Hospital Clínico Universitario de Santiago, Santiago de Compostela, Spain
| | | | | | | | | | | | - Raimon Sanmartí
- Rheumatology Department, Hospital Clínic de Barcelona and IDIBAPS, Barcelona, Spain
| | | | | | - Sílvia Bonàs-Guarch
- Barcelona Supercomputing Centre (BSC), Joint BSC-CRG-IRB Research Program in Computational Biology, Barcelona, Spain
| | - Josep Maria Mercader
- Barcelona Supercomputing Centre (BSC), Joint BSC-CRG-IRB Research Program in Computational Biology, Barcelona, Spain
| | - David Torrents
- Barcelona Supercomputing Centre (BSC), Joint BSC-CRG-IRB Research Program in Computational Biology, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Laia Codó
- Life Sciences Department, Barcelona Supercomputing Centre, Barcelona, Spain
| | - Josep Lluís Gelpí
- Life Sciences Department, Barcelona Supercomputing Centre, Barcelona, Spain
| | | | - Andrea Pluma
- Rheumatology Research Group, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Maria López-Lasanta
- Rheumatology Research Group, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Raül Tortosa
- Rheumatology Research Group, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Nuria Palau
- Rheumatology Research Group, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Devin Absher
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, USA
| | - Richard Myers
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama, USA
| | - Sara Marsal
- Rheumatology Research Group, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Antonio Julià
- Rheumatology Research Group, Vall d'Hebron Research Institute, Barcelona, Spain
| |
Collapse
|
13
|
Mead TJ, Apte SS. ADAMTS proteins in human disorders. Matrix Biol 2018; 71-72:225-239. [PMID: 29885460 DOI: 10.1016/j.matbio.2018.06.002] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 06/04/2018] [Accepted: 06/05/2018] [Indexed: 02/07/2023]
Abstract
ADAMTS proteins are a superfamily of 26 secreted molecules comprising two related, but distinct families. ADAMTS proteases are zinc metalloendopeptidases, most of whose substrates are extracellular matrix (ECM) components, whereas ADAMTS-like proteins lack a metalloprotease domain, reside in the ECM and have regulatory roles vis-à-vis ECM assembly and/or ADAMTS activity. Evolutionary conservation and expansion of ADAMTS proteins in mammals is suggestive of crucial embryologic or physiological roles in humans. Indeed, Mendelian disorders or birth defects resulting from naturally occurring ADAMTS2, ADAMTS3, ADAMTS10, ADAMTS13, ADAMTS17, ADAMTS20, ADAMTSL2 and ADAMTSL4 mutations as well as numerous phenotypes identified in genetically engineered mice have revealed ADAMTS participation in major biological pathways. Important roles have been identified in a few acquired conditions. ADAMTS5 is unequivocally implicated in pathogenesis of osteoarthritis via degradation of aggrecan, a major structural proteoglycan in cartilage. ADAMTS7 is strongly associated with coronary artery disease and promotes atherosclerosis. Autoantibodies to ADAMTS13 lead to a platelet coagulopathy, thrombotic thrombocytopenic purpura, which is similar to that resulting from ADAMTS13 mutations. ADAMTS proteins have numerous potential connections to other human disorders that were identified by genome-wide association studies. Here, we review inherited and acquired human disorders in which ADAMTS proteins participate, and discuss progress and prospects in therapeutics.
Collapse
Affiliation(s)
- Timothy J Mead
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, United States
| | - Suneel S Apte
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, United States.
| |
Collapse
|
14
|
Julià A, López-Longo FJ, Pérez Venegas JJ, Bonàs-Guarch S, Olivé À, Andreu JL, Aguirre-Zamorano MÁ, Vela P, Nolla JM, de la Fuente JLM, Zea A, Pego-Reigosa JM, Freire M, Díez E, Rodríguez-Almaraz E, Carreira P, Blanco R, Taboada VM, López-Lasanta M, Corbeto ML, Mercader JM, Torrents D, Absher D, Marsal S, Fernández-Nebro A. Genome-wide association study meta-analysis identifies five new loci for systemic lupus erythematosus. Arthritis Res Ther 2018; 20:100. [PMID: 29848360 PMCID: PMC5977506 DOI: 10.1186/s13075-018-1604-1] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 04/23/2018] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is a common systemic autoimmune disease with a complex genetic inheritance. Genome-wide association studies (GWAS) have significantly increased the number of significant loci associated with SLE risk. To date, however, established loci account for less than 30% of the disease heritability and additional risk variants have yet to be identified. Here we performed a GWAS followed by a meta-analysis to identify new genome-wide significant loci for SLE. METHODS We genotyped a cohort of 907 patients with SLE (cases) and 1524 healthy controls from Spain and performed imputation using the 1000 Genomes reference data. We tested for association using logistic regression with correction for the principal components of variation. Meta-analysis of the association results was subsequently performed on 7,110,321 variants using genetic data from a large cohort of 4036 patients with SLE and 6959 controls of Northern European ancestry. Genetic association was also tested at the pathway level after removing the effect of known risk loci using PASCAL software. RESULTS We identified five new loci associated with SLE at the genome-wide level of significance (p < 5 × 10- 8): GRB2, SMYD3, ST8SIA4, LAT2 and ARHGAP27. Pathway analysis revealed several biological processes significantly associated with SLE risk: B cell receptor signaling (p = 5.28 × 10- 6), CTLA4 co-stimulation during T cell activation (p = 3.06 × 10- 5), interleukin-4 signaling (p = 3.97 × 10- 5) and cell surface interactions at the vascular wall (p = 4.63 × 10- 5). CONCLUSIONS Our results identify five novel loci for SLE susceptibility, and biologic pathways associated via multiple low-effect-size loci.
Collapse
Affiliation(s)
- Antonio Julià
- Rheumatology Research Group, Vall d'Hebron Research Institute, 08035, Barcelona, Spain.
| | | | - José J Pérez Venegas
- Department of Rheumatology, Hospital del SAS de Jerez de la Frontera, 11407, Cádiz, Spain
| | - Silvia Bonàs-Guarch
- Barcelona Supercomputing Center. Joint BSC-CRG-IRB Research Program in Computational Biology, 08034, Barcelona, Spain
| | - Àlex Olivé
- Department of Rheumatology, Hospital Universitari Germans Trias i Pujol, 08916, Badalona, Spain
| | - José Luís Andreu
- Department of Rheumatology, Hospital Universitario Puerta de Hierro, 28222, Madrid, Spain
| | | | - Paloma Vela
- Department of Rheumatology, Hospital General Universitario de Alicante, 03010, Alicante, Spain
| | - Joan M Nolla
- Department of Rheumatology, Hospital Universitari de Bellvitge, 08907, Barcelona, Spain
| | | | - Antonio Zea
- Department of Rheumatology, Hospital Universitario Ramón y Cajal, 28034, Madrid, Spain
| | - José María Pego-Reigosa
- Department of Rheumatology, Hospital do Meixoeiro, Grupo IRIDIS, Instituto de Investigación sanitaria Galicia Sur (IISGS), 36312, Vigo, Spain
| | - Mercedes Freire
- Department of Rheumatology, Hospital Universitario A Coruña, 15006, A Coruña, Spain
| | - Elvira Díez
- Department of Rheumatology, Hospital Complejo Asistencial Universitario de León, 24071, León, Spain
| | | | - Patricia Carreira
- Department of Rheumatology, Hospital Universitario 12 de Octubre, 28041, Madrid, Spain
| | - Ricardo Blanco
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, 39008, Santander, Spain
| | - Víctor Martínez Taboada
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, 39008, Santander, Spain
| | - María López-Lasanta
- Rheumatology Research Group, Vall d'Hebron Research Institute, 08035, Barcelona, Spain
| | - Mireia López Corbeto
- Rheumatology Research Group, Vall d'Hebron Research Institute, 08035, Barcelona, Spain
| | - Josep M Mercader
- Barcelona Supercomputing Center. Joint BSC-CRG-IRB Research Program in Computational Biology, 08034, Barcelona, Spain
| | - David Torrents
- Barcelona Supercomputing Center. Joint BSC-CRG-IRB Research Program in Computational Biology, 08034, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010, Barcelona, Spain
| | - Devin Absher
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, 35806, USA
| | - Sara Marsal
- Rheumatology Research Group, Vall d'Hebron Research Institute, 08035, Barcelona, Spain.
| | - Antonio Fernández-Nebro
- Department of Rheumatology, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga, 29011, Málaga, Spain
| |
Collapse
|
15
|
Aggarwal S, Nayek A, Pradhan D, Verma R, Yadav M, Ponnusamy K, Jain AK. dbGAPs: A comprehensive database of genes and genetic markers associated with psoriasis and its subtypes. Genomics 2017; 110:S0888-7543(17)30115-5. [PMID: 29031638 DOI: 10.1016/j.ygeno.2017.10.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 10/04/2017] [Accepted: 10/10/2017] [Indexed: 01/12/2023]
Abstract
Psoriasis is a systemic hyperproliferative inflammatory skin disorder, although rarely fatal but significantly reduces quality of life. Understanding the full genetic component of the disease association may provide insight into biological pathways as well as targets and biomarkers for diagnosis, prognosis and therapy. Studies related to psoriasis associated genes and genetic markers are scattered and not easily amendable to data-mining. To alleviate difficulties, we have developed dbGAPs an integrated knowledgebase representing a gateway to psoriasis associated genomic data. The database contains annotation for 202 manually curated genes associated with psoriasis and its subtypes with cross-references. Functional enrichment of these genes, in context of Gene Ontology and pathways, provide insight into their important role in psoriasis etiology and pathogenesis. The dbGAPs interface is enriched with an interactive search engine for data retrieval along with unique customized tools for Single Nucleotide Polymorphism (SNP)/indel detection and SNP/indel annotations. dbGAPs is accessible at http://www.bmicnip.in/dbgaps/.
Collapse
Affiliation(s)
- Shweta Aggarwal
- Biomedical Informatics Centre, National Institute of Pathology - ICMR, New Delhi, India
| | - Arnab Nayek
- Biomedical Informatics Centre, National Institute of Pathology - ICMR, New Delhi, India
| | - Dibyabhaba Pradhan
- Biomedical Informatics Centre, National Institute of Pathology - ICMR, New Delhi, India
| | - Rashi Verma
- Biomedical Informatics Centre, National Institute of Pathology - ICMR, New Delhi, India
| | - Monika Yadav
- Biomedical Informatics Centre, National Institute of Pathology - ICMR, New Delhi, India
| | | | - Arun Kumar Jain
- Biomedical Informatics Centre, National Institute of Pathology - ICMR, New Delhi, India.
| |
Collapse
|
16
|
Perrotti PP, Aterido A, Fernández-Nebro A, Cañete JD, Ferrándiz C, Tornero J, Gisbert JP, Domènech E, Fernández-Gutiérrez B, Gomollón F, García-Planella E, Fernández E, Sanmartí R, Gratacós J, Martínez-Taboada VM, Rodríguez-Rodríguez L, Palau N, Tortosa R, Corbeto ML, Lasanta ML, Marsal S, Julià A. Genetic variation associated with cardiovascular risk in autoimmune diseases. PLoS One 2017; 12:e0185889. [PMID: 28982122 PMCID: PMC5628882 DOI: 10.1371/journal.pone.0185889] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 09/21/2017] [Indexed: 01/26/2023] Open
Abstract
Autoimmune diseases have a higher prevalence of cardiovascular events compared to the general population. The objective of this study was to investigate the genetic basis of cardiovascular disease (CVD) risk in autoimmunity. We analyzed genome-wide genotyping data from 6,485 patients from six autoimmune diseases that are associated with a high socio-economic impact. First, for each disease, we tested the association of established CVD risk loci. Second, we analyzed the association of autoimmune disease susceptibility loci with CVD. Finally, to identify genetic patterns associated with CVD risk, we applied the cross-phenotype meta-analysis approach (CPMA) on the genome-wide data. A total of 17 established CVD risk loci were significantly associated with CVD in the autoimmune patient cohorts. From these, four loci were found to have significantly different genetic effects across autoimmune diseases. Six autoimmune susceptibility loci were also found to be associated with CVD risk. Genome-wide CPMA analysis identified 10 genetic clusters strongly associated with CVD risk across all autoimmune diseases. Two of these clusters are highly enriched in pathways previously associated with autoimmune disease etiology (TNFα and IFNγ cytokine pathways). The results of this study support the presence of specific genetic variation associated with the increase of CVD risk observed in autoimmunity.
Collapse
Affiliation(s)
- Pedro P. Perrotti
- Rheumatology Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Adrià Aterido
- Rheumatology Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Antonio Fernández-Nebro
- UGC Reumatología, Instituto de Investigación Biomédica (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain
| | - Juan D. Cañete
- Hospital Clínic de Barcelona and IDIBAPS, Barcelona, Spain
| | | | - Jesús Tornero
- Hospital Universitario Guadalajara, Guadalajara, Spain
| | - Javier P. Gisbert
- CIBERehd, Madrid, Spain
- Hospital Universitario de la Princesa and IIS-IP, Madrid, Spain
| | - Eugeni Domènech
- Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- CIBERehd, Madrid, Spain
| | | | - Fernando Gomollón
- CIBERehd, Madrid, Spain
- Hospital Clínico Universitario, Zaragoza, Spain
| | | | | | | | | | | | | | - Núria Palau
- Rheumatology Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
| | - Raül Tortosa
- Rheumatology Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
| | - Mireia L. Corbeto
- Rheumatology Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
| | - María L. Lasanta
- Rheumatology Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
| | - Sara Marsal
- Rheumatology Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
| | - Antonio Julià
- Rheumatology Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
| | | |
Collapse
|
17
|
Uebe S, Ehrlicher M, Ekici AB, Behrens F, Böhm B, Homuth G, Schurmann C, Völker U, Jünger M, Nauck M, Völzke H, Traupe H, Krawczak M, Burkhardt H, Reis A, Hüffmeier U. Genome-wide association and targeted analysis of copy number variants with psoriatic arthritis in German patients. BMC MEDICAL GENETICS 2017; 18:92. [PMID: 28835222 PMCID: PMC5569473 DOI: 10.1186/s12881-017-0447-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 07/31/2017] [Indexed: 01/16/2023]
Abstract
Background Psoriatic Arthritis (PsA) is a chronic inflammatory disease of the joints. PsA is etiologically complex, and 11 susceptibility loci have been identified so far. Most of these overlap with loci associated with psoriasis vulgaris (PsV), the most common psoriatic skin manifestation which is also frequently seen in PsA patients. In addition, two copy number variants (CNVs) are associated with PsV, one of which, located within the LCE3 gene cluster, is also associated with PsA. Finally, an intergenic deletion has been reported as a PsA-specific CNV. Methods We performed a genome-wide association study (GWAS) of CNVs in PsA and assessed the contribution to disease risk by CNVs at known psoriasis susceptibility loci. Results After stringent quality assessment and validation of CNVs of the GWAS with an alternative quantitative method, two significantly associated CNVs remained, one near UXS1, the other one at the TRB locus. However, MLPA analysis did not confirm the CN state in ~1/3 of individuals, and an analysis of an independent case-control-study failed to confirm the initial associations. Furthermore, detailed PCR-based analysis of the sequence at TRB revealed the existence of a more complex genomic sequence most accurately represented by freeze hg18 which accordingly failed to confirm the hg19 sequence. Only rare CNVs were detected at psoriasis susceptibility loci. At three of 12 susceptibility loci with CNVs (CSMD1, IL12B, RYR2), CN variability was confirmed independently by MLPA. Overall, the rate of CNV confirmation by MLPA was strongly dependent upon CNV type, CNV size and the number of array markers involved in a CNV. Conclusion Although we identified PsA associations at several loci and confirmed that the common CNVs at these sites were real, ~1/3 of the common CNV states could not be reproduced. Furthermore, replication analysis failed to confirm the original association. Furthermore, SNP array-based analyses of CNVs were found to be more reliable for deletions than duplications, independent of the respective CNV allele frequency. CNVs are thus good candidate disease variants, while the methods to detect them should be applied cautiously and reproduced by an independent method. Electronic supplementary material The online version of this article (doi:10.1186/s12881-017-0447-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Steffen Uebe
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 10, 91054, Erlangen, Germany
| | - Maria Ehrlicher
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 10, 91054, Erlangen, Germany
| | - Arif Bülent Ekici
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 10, 91054, Erlangen, Germany
| | - Frank Behrens
- Division of Rheumatology and IME Fraunhofer Project Group Translational Medicine & Pharmacology, Goethe University, Frankfurt/Main, Germany
| | - Beate Böhm
- Division of Rheumatology and IME Fraunhofer Project Group Translational Medicine & Pharmacology, Goethe University, Frankfurt/Main, Germany
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine and Ernst-Moritz-Arndt University Greifswald, Greifswald, Germany
| | - Claudia Schurmann
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine and Ernst-Moritz-Arndt University Greifswald, Greifswald, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine and Ernst-Moritz-Arndt University Greifswald, Greifswald, Germany
| | - Michael Jünger
- Clinic of Dermatology, University of Greifswald, Greifswald, Germany
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University of Greifswald, Greifswald, Germany
| | - Henry Völzke
- Institute for Community Medicine, University of Greifswald, Greifswald, Germany
| | - Heiko Traupe
- Department of Dermatology, University of Münster, Münster, Germany
| | - Michael Krawczak
- Institute for Medical Informatics and Statistics, Christian-Albrechts University Kiel, Kiel, Germany
| | - Harald Burkhardt
- Division of Rheumatology and IME Fraunhofer Project Group Translational Medicine & Pharmacology, Goethe University, Frankfurt/Main, Germany
| | - André Reis
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 10, 91054, Erlangen, Germany
| | - Ulrike Hüffmeier
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 10, 91054, Erlangen, Germany.
| |
Collapse
|