1
|
Tanimura K, Aldrich MC, Jaworski J, Xing J, Okawa S, Chandra D, Nouraie SM, Nyunoya T. Identifying a Genetic Link Between Lung Function and Psoriasis. Ann Hum Genet 2025; 89:89-95. [PMID: 39718377 PMCID: PMC11982659 DOI: 10.1111/ahg.12587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 12/03/2024] [Accepted: 12/07/2024] [Indexed: 12/25/2024]
Abstract
INTRODUCTION The common genetic underpinnings of psoriasis and pulmonary comorbidities have yet to be explored. MATERIAL AND METHODS In this cross-sectional study, we investigated the single-nucleotide polymorphisms (SNPs) associated with psoriasis and their relationship with pulmonary function using data from the UK Biobank (UKBB) and the Vanderbilt University Medical Center Biobank (BioVU). RESULTS Out of the 63 psoriasis-associated SNPs identified in previous genome-wide association studies within the European population, we successfully identified 53 SNPs, including proxy SNPs in UKBB database. Following adjustments using age and sex, 31 SNPs displayed statistically significant associations with psoriasis. Among these, 16 SNPs exhibited significant associations with forced expiratory volume in 1 s (FEV1), 14 with forced vital capacity (FVC), and 5 with the FEV1/FVC ratio in the UKBB. In the validation analysis using the BioVU database, 27 of the 31 psoriasis-associated SNPs were available for examination. Notably, the minor allele of SNP rs8016947 was confirmed to be significant, indicating a reduced risk for psoriasis and improved FEV1. Similarly, the minor alleles of SNPs rs17716942 and rs8016947 were associated with a reduced risk of psoriasis and enhanced FVC. However, none of the 5 SNPs significantly associated with the FEV1/FVC ratio in the UKBB displayed significance in the BioVU dataset. CONCLUSION This study has unveiled genetic variants that bridge the realms of psoriasis and lung function. The genes associated with these variants, including IFIH1, Grancalcin gene (GCA), and NFKB inhibitor alpha gene (NFKBIA), regulate innate immune responses, which suggests that immunodysregulation, a central element in psoriasis pathogenesis, may also impact lung function, alluding to a "skin-lung axis."
Collapse
Affiliation(s)
- Kazuya Tanimura
- Department of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of Respiratory MedicineNara Medical UniversityKashiharaNaraJapan
| | - Melinda C. Aldrich
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - James Jaworski
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Jinchuan Xing
- Department of GeneticsHuman Genetic Institute of New Jersey, Rutgers, the State University of New JerseyPiscatawayNew JerseyUSA
| | - Satoshi Okawa
- Department of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Divay Chandra
- Department of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Seyed M. Nouraie
- Department of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Toru Nyunoya
- Department of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Medical Specialty Service LineVeterans Affairs Pittsburgh Healthcare SystemPittsburghPennsylvaniaUSA
| |
Collapse
|
2
|
Perez-Chada LM, Elman S, Villa-Ruiz C, Armstrong AW, Gottlieb AB, Merola JF. Psoriatic arthritis: A comprehensive review for the dermatologist part I: Epidemiology, comorbidities, pathogenesis, and diagnosis. J Am Acad Dermatol 2025; 92:969-982. [PMID: 38857765 DOI: 10.1016/j.jaad.2024.03.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 02/22/2024] [Accepted: 03/08/2024] [Indexed: 06/12/2024]
Abstract
Psoriatic arthritis (PsA) is an inflammatory seronegative arthritis strongly associated with psoriasis. Recognition of the clinical features of PsA is critical, as delayed detection and untreated disease may result in irreparable joint damage, impaired physical function, and a significantly reduced quality of life. Dermatologists are poised for the early detection of PsA, as psoriasis predates its development in as many as 80% of patients. In an effort to further acquaint dermatologists with PsA, this review provides a detailed overview, emphasizing its epidemiology, comorbidities, etiopathogenesis, and diagnostic features.
Collapse
Affiliation(s)
- Lourdes M Perez-Chada
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Scott Elman
- Dr. Phillip Frost Department of Dermatology & Cutaneous Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida
| | - Camila Villa-Ruiz
- Department of Dermatology, Boston University School of Medicine, Boston, Massachusetts
| | - April W Armstrong
- Department of Dermatology, University of California, Los Angeles, Los Angeles, California
| | - Alice B Gottlieb
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Joseph F Merola
- Department of Dermatology and Department of Medicine, Division of Rheumatology and O'Donnell School of Public Health, UT Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
3
|
Bonomo MG, D’Angelo S, Picerno V, Carriero A, Salzano G. Recent Advances in Gut Microbiota in Psoriatic Arthritis. Nutrients 2025; 17:1323. [PMID: 40284188 PMCID: PMC12030176 DOI: 10.3390/nu17081323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/04/2025] [Accepted: 04/08/2025] [Indexed: 04/29/2025] Open
Abstract
Psoriatic arthritis (PsA) is a chronic inflammatory disease characterized by joint inflammation and skin lesions. Recent research has underscored the critical role of gut microbiota-comprising bacteria, fungi, viruses, and archaea-in the pathogenesis and progression of PsA. This narrative review synthesizes the latest findings on the influence of gut microbiota on PsA, focusing on mechanisms such as immune modulation, microbial dysbiosis, the gut-joint axis, and its impact on treatment. Advances in high-throughput sequencing and metagenomics have revealed distinct microbial profiles associated with PsA. Studies show that individuals with PsA have a unique gut microbiota composition, differing significantly from healthy controls. Alterations in the abundance of specific bacterial taxa, including a decrease in beneficial bacteria and an increase in potentially pathogenic microbes, contribute to systemic inflammation by affecting the intestinal barrier and promoting immune responses. This review explores the impact of various factors on gut microbiota composition, including age, hygiene, comorbidities, and medication use. Additionally, it highlights the role of diet, probiotics, and fecal microbiota transplantation as promising strategies to modulate gut microbiota and alleviate PsA symptoms. The gut-skin-joint axis concept illustrates how gut microbiota influences not only gastrointestinal health but also skin and joint inflammation. Understanding the complex interplay between gut microbiota and PsA could lead to novel, microbiome-based therapeutic approaches. These insights offer hope for improved patient outcomes through targeted manipulation of the gut microbiota, enhancing both diagnosis and treatment strategies for PsA.
Collapse
Affiliation(s)
- Maria Grazia Bonomo
- Department of Health Sciences, University of Basilicata, Viale dell’ Ateneo Lucano 10, 85100 Potenza, Italy; (S.D.); (G.S.)
| | - Salvatore D’Angelo
- Department of Health Sciences, University of Basilicata, Viale dell’ Ateneo Lucano 10, 85100 Potenza, Italy; (S.D.); (G.S.)
- Rheumatology Department of Lucania, San Carlo Hospital of Potenza, Via Potito Petrone, 85100 Potenza, Italy; (V.P.); (A.C.)
| | - Valentina Picerno
- Rheumatology Department of Lucania, San Carlo Hospital of Potenza, Via Potito Petrone, 85100 Potenza, Italy; (V.P.); (A.C.)
| | - Antonio Carriero
- Rheumatology Department of Lucania, San Carlo Hospital of Potenza, Via Potito Petrone, 85100 Potenza, Italy; (V.P.); (A.C.)
| | - Giovanni Salzano
- Department of Health Sciences, University of Basilicata, Viale dell’ Ateneo Lucano 10, 85100 Potenza, Italy; (S.D.); (G.S.)
| |
Collapse
|
4
|
Zabotti A, Aydin SZ, David P, Di Matteo A, McGonagle D. Delineating inflammatory from non-inflammatory mechanisms for therapy optimization in psoriatic arthritis. Nat Rev Rheumatol 2025; 21:237-248. [PMID: 40075177 DOI: 10.1038/s41584-025-01229-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2025] [Indexed: 03/14/2025]
Abstract
Psoriatic arthritis (PsA) is anatomically much more heterogeneous than rheumatoid arthritis, as, beyond synovitis, it often also involves enthesitis, peritendinitis, tenosynovitis, osteitis and periostitis. This heterogeneity currently precludes a gold standard for objectively defining resolution of inflammation following treatment, with enthesitis posing a particular challenge. Despite these difficulties, we apply lessons learned from rheumatoid arthritis to describe how patients with PsA and an inadequate response to therapy can be designated within two patient subgroups, characterized by persistent inflammatory PsA (PIPsA) and non-inflammatory PsA (NIPsA), respectively. The NIPsA phenotype is defined by the lack of ongoing joint inflammation, as confirmed through clinical assessment and imaging, along with normalized inflammatory marker levels. NIPsA might be associated with obesity, biomechanical-related pain, osteoarthritis, fibromyalgia, secondary post-inflammatory damage and central pain mechanisms. In this article, we frame PsA composite outcomes measures in relationship to the PIPsA and NIPsA phenotypes and propose that this approach might help to minimize unnecessary or ineffective cycling of PsA therapy in patients who acquire dominant non-inflammatory mechanisms and might also inform future trial design.
Collapse
Affiliation(s)
- Alen Zabotti
- Section of Musculoskeletal Disease, NIHR Leeds Musculoskeletal Biomedical Research Centre, Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Chapel Allerton Hospital, Leeds, UK
- Division of Rheumatology, Azienda Sanitaria Universitaria del Friuli Centrale, Santa Maria della Misericordia Hospital, Udine, Italy
| | - Sibel Zehra Aydin
- University of Ottawa, The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Paula David
- Section of Musculoskeletal Disease, NIHR Leeds Musculoskeletal Biomedical Research Centre, Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Chapel Allerton Hospital, Leeds, UK
- Department of Internal Medicine B & Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel. Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Andrea Di Matteo
- Section of Musculoskeletal Disease, NIHR Leeds Musculoskeletal Biomedical Research Centre, Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Chapel Allerton Hospital, Leeds, UK
| | - Dennis McGonagle
- Section of Musculoskeletal Disease, NIHR Leeds Musculoskeletal Biomedical Research Centre, Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Chapel Allerton Hospital, Leeds, UK.
| |
Collapse
|
5
|
Saeed F, Adamopoulos IE. Pathogenesis of psoriatic arthritis: new insights from a bone marrow perspective. Curr Opin Rheumatol 2025; 37:136-141. [PMID: 39470182 PMCID: PMC11779588 DOI: 10.1097/bor.0000000000001064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
PURPOSE OF REVIEW Psoriatic arthritis is an immune-mediated disease that primarily affects the skin and joints. It falls under the umbrella term of rheumatic diseases, which describes a group of closely related yet distinct disorders with many common underlying molecular pathways. Despite the distinct clinical manifestation of each disorder, the shared therapeutic strategies attest to the commonality of cellular and molecular underpinnings. Herein we provide a concise yet comprehensive overview of the interleukin (IL)-23/IL-17 axis and its involvement in mechanistic pathways leading to the pathogenesis of this dual skin and joint clinical manifestation which is characteristic of psoriatic arthritis and other rheumatic diseases. RECENT FINDINGS The interconnection between activated innate immune cells and adaptive immunity has transformed current thinking to include other organs such as the bone marrow as potential tissue of disease origin. A plethora of animal models and genetic studies converge on the critical role of IL-23/IL-17 axis, and highlight the importance of myeloid cell activation as common pathways between autoinflammatory and autoimmune diseases and chronic inflammation. These findings underscore the intricate immune mechanisms involved in inflammatory arthritis and highlight molecular mechanisms in disease pathogenesis. SUMMARY These insights pave the way for the development of novel diagnostic and therapeutic strategies, with a focus on translating these findings into improved clinical practice.
Collapse
Affiliation(s)
- Fatima Saeed
- Department of Rheumatology and Clinical Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Iannis E. Adamopoulos
- Department of Rheumatology and Clinical Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| |
Collapse
|
6
|
Dilek G, Kalcik Unan M, Nas K. Immune response and cytokine pathways in psoriatic arthritis: A systematic review. Arch Rheumatol 2025; 40:144-156. [PMID: 40264485 PMCID: PMC12010266 DOI: 10.46497/archrheumatol.2025.10934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 01/03/2025] [Indexed: 04/24/2025] Open
Abstract
Objectives This review aims to focus on the role of innate and adaptive immune system cells and their molecular signaling pathways in the pathophysiology of clinical phenotypes of psoriatic arthritis (PsA). Materials and methods A systematic literature search was conducted in the PubMed database using the key words "psoriasis," "psoriatic arthritis," "pathogenesis," "adaptive immune system," "pathophysiology," "bone and cartilage damage," and "cytokine pathways." Results Clinical studies and in vitro studies on adaptive and innate immune system cells and mediators that cause activation of these cells in the pathogenesis of PsA were examined. The role of cytokine pathways affecting the pathogenesis of PsA on the most common clinical phenotypes of the disease were explained in detail. Conclusion In this article, we reviewed the cytokine pathways that may contribute to the immunological pathogenesis of psoriatic arthritis. We believe that this review will contribute to a better understanding of the pathogenesis of the clinical phenotypes of the disease and to disease management.
Collapse
Affiliation(s)
- Gamze Dilek
- Abant İzzet Baysal University Training and Research Hospital, Rheumatology Clinic, Bolu, Türkiye
| | - Mehtap Kalcik Unan
- Department of Physical Medicine and Rehabilitation, Division of Rheumatology and Immunology, Sakarya University School of Medicine, Sakarya, Türkiye
| | - Kemal Nas
- Department of Physical Medicine and Rehabilitation, Division of Rheumatology and Immunology, Sakarya University School of Medicine, Sakarya, Türkiye
| |
Collapse
|
7
|
Maaloul M, Charfi A, Feki A, Gassara Z, Hakim F, Gaddour L, Fourati H, Baklouti S, Kamoun A, Mahfoudh N. Association study of the HLA class I system with psoriatic arthritis in Southern Tunisia: a case-control study. Clin Rheumatol 2025; 44:707-718. [PMID: 39715961 DOI: 10.1007/s10067-024-07290-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 12/25/2024]
Abstract
INTRODUCTION/OBJECTIVES Psoriatic arthritis (PsA) is a chronic inflammatory rheumatism belonging to the spondyloarthritis family. It is a multifactorial disease whose genetic determinism is still poorly understood. It is favored by environmental factors in genetically predisposed individuals. This study aims to investigate the association of HLA-A, HLA-B, and HLA-C alleles with PsA and its various manifestations in patients from Southern Tunisia. PATIENTS AND METHODS A case-control association study was conducted involving 48 PsA patients and 123 controls. HLA-A and HLA-B typing was performed using microlymphocytotoxicity complement-dependent assays, and HLA-C typing was done using polymerase chain reaction-sequence specific primer. RESULTS Positive associations were confirmed for HLA-B27 and -C*06 alleles with PsA in our Southern Tunisian population with a more pronounced association in cases of -C*06 homozygosity. The HLA-B*27-C*02 and HLA-B*50-C*06 haplotypes were significantly more frequent in PsA patients, whereas the HLA-B*35-C*04 haplotype was negatively associated with PsA. Specific HLA alleles were linked to disease manifestations: -C04 with female sex, -B27 with familial spondyloarthritis, and -B17 with sporadic PsA. Cervical spine involvement was associated with -C02, and hip involvement with -B35 and -C02. Uveitis was linked to -C02 and -B27. A negative association was observed between a BASFI score > 4 and the presence of -B45 and -C07. CONCLUSION The HLA class I system contributes to PsA susceptibility in the Southern Tunisian population. Key Points • This is the first study exploring the association between HLA class I alleles and psoriatic arthritis in Southern Tunisia. • HLA-B27 and -C06 alleles are strongly associated with PsA, with a more pronounced effect in -C06 homozygosity. • Specific HLA alleles are linked to clinical manifestations: -B27 is associated with familial PsA, -B17 with sporadic PsA, and -B35 with hip involvement. • These findings provide insights into the genetic contribution to PsA pathophysiology in Southern Tunisia and highlight the role of HLA alleles in specific disease features.
Collapse
Affiliation(s)
- Mariem Maaloul
- Immunology and Histocompatibility Department, Hedi Chaker University Hospital, Sfax, Tunisia.
| | - Aida Charfi
- Immunology and Histocompatibility Department, Hedi Chaker University Hospital, Sfax, Tunisia
| | - Afef Feki
- Rheumatology Department, Hedi Chaker University Hospital, Sfax, Tunisia
| | - Zouhour Gassara
- Rheumatology Department, Hedi Chaker University Hospital, Sfax, Tunisia
| | - Feiza Hakim
- Immunology and Histocompatibility Department, Hedi Chaker University Hospital, Sfax, Tunisia
| | - Lilia Gaddour
- Immunology and Histocompatibility Department, Hedi Chaker University Hospital, Sfax, Tunisia
| | - Hela Fourati
- Rheumatology Department, Hedi Chaker University Hospital, Sfax, Tunisia
| | - Sofien Baklouti
- Rheumatology Department, Hedi Chaker University Hospital, Sfax, Tunisia
| | - Arwa Kamoun
- Immunology and Histocompatibility Department, Hedi Chaker University Hospital, Sfax, Tunisia
| | - Nadia Mahfoudh
- Immunology and Histocompatibility Department, Hedi Chaker University Hospital, Sfax, Tunisia
| |
Collapse
|
8
|
Stadler M, Zhao SS, Bowes J. A review of the advances in understanding the genetic basis of spondylarthritis and emerging clinical benefit. Best Pract Res Clin Rheumatol 2024; 38:101982. [PMID: 39223061 DOI: 10.1016/j.berh.2024.101982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 09/04/2024]
Abstract
Spondyloarthropathies (SpA), including ankylosing spondylitis (AS) and psoriatic arthritis (PsA), have been shown to have a substantial genetic predisposition based on heritability estimates derived from family studies and genome-wide association studies (GWAS). GWAS have uncovered numerous genetic loci associated with susceptibility to SpA, with significant associations to human leukocyte antigen (HLA) genes, which are major genetic risk factors for both AS and PsA. Specific loci differentiating PsA from cutaneous-only psoriasis have been identified, though these remain limited. Further research with larger sample sizes is necessary to identify more PsA-specific genetic markers. Current research focuses on translating these genetic insights into clinical applications. For example, polygenic risk scores are showing promise for the classification of disease risk and diagnosis and future research should focus on refining these risk assessment tools to improve clinical outcomes for individuals with SpA. Addressing these challenges will help integrate genetic testing into patients care and impact clinical practice.
Collapse
Affiliation(s)
- Michael Stadler
- The Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Sizheng Steven Zhao
- The Centre for Epidemiology Versus Arthritis, Centre for Musculoskeletal Research, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - John Bowes
- The Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK.
| |
Collapse
|
9
|
Caso F, Costa L, Megna M, Cascone M, Maione F, Giacomelli R, Scarpa R, Ruscitti P. Early psoriatic arthritis: clinical and therapeutic challenges. Expert Opin Investig Drugs 2024; 33:945-965. [PMID: 39041193 DOI: 10.1080/13543784.2024.2383421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
INTRODUCTION Psoriatic arthritis (PsA) is a chronic immunoinflammatory disease of the enthesis and adjacent synovium, skin, and nail, which early diagnosis may be crucial for starting a prompt therapeutic intervention. Theoretically, early treatment offers the advantage of acting on the reduction of the articular damage progression since initial phases of the disease. AREAS COVERED This review explores the challenges of clinical-diagnostic aspects and the underlying pathophysiology of early PsA phases, as well as the evidence evaluating the impact of early intervention on disease outcomes. EXPERT OPINION Main instruments for early PsA diagnosis include recognizing synovial-entheseal inflammatory signs at onset, improving screening PsA high-risk subjects, and increasing disease knowledge of physicians and patients with psoriasis or familial history. PsA continues to significantly impact on the Quality of Life of patients affected by the disease, making necessary to deeply study clinical manifestations, risk factors, and underlying immunoinflammatory mechanisms, as well as to identify biomarkers for early identification. Additionally, it remains a need to increase more evidence on understanding how early treatment of PsA and of psoriasis might influence the course of the disease.
Collapse
Affiliation(s)
- Francesco Caso
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Luisa Costa
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Matteo Megna
- Section of Dermatology - Department of Clinical Medicine and Surgery, University of Naples Federico II, Napoli, Italy
| | - Mario Cascone
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Francesco Maione
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Roberto Giacomelli
- Research and Clinical Unit of immunorheumatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Rheumatology, Immunology and Clinical Medicine Unit, Department of Medicine, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Raffaele Scarpa
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Piero Ruscitti
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
10
|
Worth C, Al-Mossawi MH, Macdonald J, Fisher BA, Chan A, Sengupta R, Packham J, Gaffney K, Gullick N, Cook JA, Corn TH, Teh J, Machado PM, Taylor PC, Bowness P. Granulocyte-macrophage colony-stimulating factor neutralisation in patients with axial spondyloarthritis in the UK (NAMASTE): a randomised, double-blind, placebo-controlled, phase 2 trial. THE LANCET. RHEUMATOLOGY 2024; 6:e537-e545. [PMID: 38942047 DOI: 10.1016/s2665-9913(24)00099-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 03/15/2024] [Accepted: 04/15/2024] [Indexed: 06/30/2024]
Abstract
BACKGROUND Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a proinflammatory cytokine overproduced in several inflammatory and autoimmune diseases, including axial spondyloarthritis. Namilumab is a human IgG1 monoclonal anti-GM-CSF antibody that potently neutralises human GM-CSF. We aimed to assess the efficacy of namilumab in participants with moderate-to-severe active axial spondyloarthritis. METHODS This proof-of-concept, randomised, double-blind, placebo-controlled, phase 2, Bayesian (NAMASTE) trial was done at nine hospitals in the UK. Participants aged 18-75 years with axial spondyloarthritis, meeting the Assessment in SpondyloArthritis international Society (ASAS) criteria and the ASAS-defined MRI criteria, with active disease as defined by a Bath Ankylosing Spondylitis Disease Activity Index (BASDAI), were eligible. Those who had inadequately responded or had intolerance to previous treatment with an anti-TNF agent were included. Participants were randomly assigned (6:1) to receive subcutaneous namilumab 150 mg or placebo at weeks 0, 2, 6, and 10. Participants, site staff (except pharmacy staff), and central study staff were masked to treatment assignment. The primary endpoint was the proportion of participants who had an ASAS ≥20% improvement (ASAS20) clinical response at week 12 in the full analysis set (all randomly assigned participants). This trial is registered with ClinicalTrials.gov (NCT03622658). FINDINGS From Sept 6, 2018, to July 25, 2019, 60 patients with moderate-to-severe active axial spondyloarthritis were assessed for eligibility and 42 were randomly assigned to receive namilumab (n=36) or placebo (n=six). The mean age of participants was 39·5 years (SD 13·3), 17 were women, 25 were men, 39 were White, and seven had previously received anti-TNF therapy. The primary endpoint was not met. At week 12, the proportion of patients who had an ASAS20 clinical response was lower in the namilumab group (14 of 36) than in the placebo group (three of six; estimated between-group difference 6·8%). The Bayesian posterior probability η was 0·72 (>0·927 suggests high clinical significance). The rates of any treatment-emergent adverse events in the namilumab group were similar to those in the placebo group (31 vs five). INTERPRETATION Namilumab did not show efficacy compared with placebo in patients with active axial spondyloarthritis, but the treatment was generally well tolerated. FUNDING Izana Bioscience, NIHR Oxford Biomedical Research Centre (BRC), NIHR Birmingham BRC, and Clinical Research Facility.
Collapse
Affiliation(s)
- Claudia Worth
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK.
| | - M Hussein Al-Mossawi
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Joanne Macdonald
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Benjamin A Fisher
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK; National Institute for Health Research (NIHR), Birmingham Biomedical Research Centre, Department of Rheumatology, University Hospitals Birmingham, NHS Foundation Trust, Birmingham, UK
| | - Antoni Chan
- Royal Berkshire, NHS Foundation Trust, Reading, UK
| | | | | | - Karl Gaffney
- Norfolk and Norwich University Hospitals, NHS Foundation Trust, Norwich, UK
| | - Nicola Gullick
- University Hospitals Coventry and Warwickshire, Warwick Medical School, University of Warwick, Warwick, UK
| | - Jonathan A Cook
- Centre for Statistics in Medicine, University of Oxford, Oxford, UK
| | - Tim H Corn
- Asclepius Consulting (Izana Bioscience), London, UK
| | - James Teh
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Pedro M Machado
- Centre for Rheumatology and Department of Neuromuscular Diseases, University College London, London, UK; NIHR, University College London Hospitals, Biomedical Research Centre, NHS Foundation Trust, London, UK; Department of Rheumatology, Northwick Park Hospital, London North West University Healthcare, NHS Trust, London, UK
| | - Peter C Taylor
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Paul Bowness
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| |
Collapse
|
11
|
Zhou G, Gan L, Zhao B, Fang F, Liu H, Chen X, Huang J. Adding salt to foods and risk of psoriasis: A prospective cohort study. J Autoimmun 2024; 147:103259. [PMID: 38823158 DOI: 10.1016/j.jaut.2024.103259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/26/2024] [Accepted: 05/16/2024] [Indexed: 06/03/2024]
Abstract
BACKGROUND High salt intake may play a critical role in the etiology of psoriasis. Yet, evidence on the association of high salt intake with risk of psoriasis is limited. OBJECTIVE To estimate the association between frequency of adding salt to foods and risk of psoriasis. METHODS We conducted a prospective cohort study of 433,788 participants from the UK Biobank. Hazard ratios (HRs) and their 95 % confidence intervals (CIs) for risk of psoriasis in relation to frequency of adding salt to foods were estimated using multivariable Cox proportional hazards models. We further evaluated the joint association of adding salt to foods and genetic susceptibility with risk of psoriasis. We conducted a mediation analysis to assess how much of the effect of adding salt to foods on risk of psoriasis was mediated through several selected mediators. RESULTS During a median of 14.0 years of follow-up, 4279 incident cases of psoriasis were identified. In the multivariable-adjusted model, a higher frequency of adding salt to foods was significantly associated with an increased risk of psoriasis ("always" versus "never/rarely" adding salt to foods, HR = 1.25, 95 % CI: 1.10, 1.41). The observed positive association was generally similar across subgroups. In the joint association analysis, we observed that participants with a high genetic risk (above the second tertile) and the highest frequency of adding salt to foods experienced 149 % higher risk of psoriasis, when compared with participants with a low genetic risk (below the first tertile) and the lowest frequency of adding salt to foods (HR = 2.49, 95 % CI: 2.05, 3.02). Mediation analysis revealed that 1.8 %-3.2 % of the positive association between frequency of adding salt and risk of psoriasis was statistically significantly mediated by obesity and inflammatory biomarkers such as C-reactive protein and systemic immune-inflammation index (all P values < 0.004). CONCLUSIONS Our study demonstrated a positive association between frequency of adding salt to foods and risk of psoriasis. The positive association was independent of multiple other risk factors, and may be partially mediated through obesity and inflammation.
Collapse
Affiliation(s)
- Guowei Zhou
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Clinical Research Center for Cancer Immunotherapy, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Engineering Laboratory of Medical Big Data Application Technology (Central South University), Changsha, China; Furong Laboratory, Changsha, China
| | - Lu Gan
- Furong Laboratory, Changsha, China; National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China; Xiangya School of Public Health, Central South University, Changsha, China; CSU-Sinocare Research Center for Nutrition and Metabolic Health, Changsha, China
| | - Bin Zhao
- Furong Laboratory, Changsha, China; National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China; Xiangya School of Public Health, Central South University, Changsha, China; CSU-Sinocare Research Center for Nutrition and Metabolic Health, Changsha, China
| | - Fang Fang
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Hong Liu
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Clinical Research Center for Cancer Immunotherapy, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Engineering Laboratory of Medical Big Data Application Technology (Central South University), Changsha, China; Furong Laboratory, Changsha, China.
| | - Xiang Chen
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Clinical Research Center for Cancer Immunotherapy, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Engineering Laboratory of Medical Big Data Application Technology (Central South University), Changsha, China; Furong Laboratory, Changsha, China.
| | - Jiaqi Huang
- Furong Laboratory, Changsha, China; National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China; Xiangya School of Public Health, Central South University, Changsha, China; CSU-Sinocare Research Center for Nutrition and Metabolic Health, Changsha, China.
| |
Collapse
|
12
|
Wu A, Scher JU, Ogdie A, Ritchlin C, Merola JF. Prevention of Psoriatic Arthritis: The Need for Prospective Studies. Dermatol Clin 2024; 42:429-438. [PMID: 38796274 DOI: 10.1016/j.det.2024.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2024]
Abstract
Psoriatic arthritis (PsA) is a systemic chronic inflammatory disease that develops in up to 30% of patients with psoriasis. Mixed data variably support the potential ability to "prevent" and/or delay PsA through use of systemic therapies in psoriasis patients. Though intriguing, almost all of these studies are retrospective in nature, and hold substantial limitations and potential biases that challenge the ability to meaningfully interpretation their results. Thus, the authors believe prospective observational and interventional studies are crucial to understanding our ability to truly modify the transition from psoriasis to psoriatic arthritis and delay or prevent PsA onset.
Collapse
Affiliation(s)
- Alexander Wu
- Department of Dermatology, UT Southwestern Medical Center, 5939 Harry Hines Boulevard 4th Floor, Suite 100, Dallas, TX 75390, USA; University of Texas Southwestern Medical School, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Jose U Scher
- Department of Medicine at NYU Grossman School of Medicine, 333 East 38th Street, 4th Floor, New York, NY 10016, USA
| | - Alexis Ogdie
- Department of Medicine (Rheumatology) and Epidemiology, Hospital of the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Christopher Ritchlin
- Department of Medicine, University of Rochester Medical Center, 400 Red Creek Drive, Suite 200, Rochester, NY 14623, USA
| | - Joseph F Merola
- Department of Dermatology, UT Southwestern Medical Center, 5939 Harry Hines Boulevard 4th Floor, Suite 100, Dallas, TX 75390, USA; Division of Rheumatology, Department of Internal Medicine, University of Texas Southwestern Medical Center; UT Southwestern Medical School and O'Donnell School of Public Health, 5939 Harry Hines Boulevard, Dallas, TX 75390, USA.
| |
Collapse
|
13
|
Assarsson M, Söderman J, Seifert O. Significant Correlation Between Cutaneous Abundance of Streptococcus and Psoriasis Severity in Patients with FBXL19 Gene Variants. Acta Derm Venereol 2024; 104:adv34892. [PMID: 38898675 PMCID: PMC11210493 DOI: 10.2340/actadv.v104.34892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
Psoriasis results from both genetic predisposition and environmental triggers, such as Streptococcal infections. This study aimed to explore the correlation between the abundance of the Streptococcus genus on the skin and psoriasis severity in individuals carrying specific psoriasis-associated genetic variants. Studying 39 chronic plaque psoriasis patients, the elbow skin microbiome and 49 psoriasis-related single nucleotide polymorphisms (SNPs) were analysed using a MiSeq instrument for 16S rDNA sequencing, and CLC Genomic Workbench for processing and analysis. Through multivariate linear regression analysis, a positive correlation was found between Streptococcus genus abundance and psoriasis severity in patients with certain FBXL19 gene-related heterozygous SNPs (rs12924903, rs10782001, rs12445568). Conversely, a negative association was observed in patients with homozygous genotypes. Moreover, we identified an association between Streptococcus abundance and psoriasis severity in patients with genetic variants related to IL-22, ERAP1, NOS2, and ILF3. This is the first study highlighting a positive association between Streptococcus skin colonization and psoriasis severity in patients with heterozygous genotypes within the FBXL19 gene region. FXBL19 targets the IL-33/IL1RL1 axis, crucial in infectious diseases and innate immunity promotion. These novel results suggests an intricate interaction among host genetics, Streptococcus skin colonization, and psoriasis inflammation, offering potential avenues for novel treatment approaches.
Collapse
Affiliation(s)
- Malin Assarsson
- Department of Biomedical and Clinical Sciences, Faculty of Health Sciences, Linköping University, Linköping, Sweden; Division of Dermatology and Venereology, Region Jönköping County, Jönköping, Sweden.
| | - Jan Söderman
- Department of Biomedical and Clinical Sciences, Faculty of Health Sciences, Linköping University, Linköping, Sweden; Laboratory Medicine, Region Jönköping County, Jönköping, Sweden
| | - Oliver Seifert
- Department of Biomedical and Clinical Sciences, Faculty of Health Sciences, Linköping University, Linköping, Sweden; Division of Dermatology and Venereology, Region Jönköping County, Jönköping, Sweden
| |
Collapse
|
14
|
Gao S, Song H. Differences between psoriatic arthritis and psoriasis in multi-omics. Arch Dermatol Res 2024; 316:217. [PMID: 38787526 DOI: 10.1007/s00403-024-03018-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 04/08/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024]
Abstract
We aim to systemically review the genomics, transcriptomics, epigenetics, proteomics, metabonomics and microbiota of psoriatic arthritis and psoriasis, illustrating the differences of these two diseases, broadening our understanding of the pathogenesis of them and providing important clues for valuable biomarkers of earlier diagnosis and treatments. To our knowledge, this is the first study that combine all omics studies from genomics to microbiota and may serve as a reference for future studies to identify the key underlying pathways in psoriatic arthritis.
Collapse
Affiliation(s)
- Siming Gao
- Department of Rheumatology, Beijing Jishuitan Hospital, Guizhou Hospital, Guiyang, China
- Department of Rheumatology, Beijing Jishuitan Hospital, Capital Medical University, No.31, Xin Jie Kou East Street, Xicheng District, Beijing, 100035, China
| | - Hui Song
- Department of Rheumatology, Beijing Jishuitan Hospital, Capital Medical University, No.31, Xin Jie Kou East Street, Xicheng District, Beijing, 100035, China.
| |
Collapse
|
15
|
Zalesak M, Danisovic L, Harsanyi S. Psoriasis and Psoriatic Arthritis-Associated Genes, Cytokines, and Human Leukocyte Antigens. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:815. [PMID: 38792999 PMCID: PMC11123327 DOI: 10.3390/medicina60050815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024]
Abstract
In recent years, research has intensified in exploring the genetic basis of psoriasis (PsO) and psoriatic arthritis (PsA). Genome-wide association studies (GWASs), including tools like ImmunoChip, have significantly deepened our understanding of disease mechanisms by pinpointing risk-associated genetic loci. These efforts have elucidated biological pathways involved in PsO pathogenesis, particularly those related to the innate immune system, antigen presentation, and adaptive immune responses. Specific genetic loci, such as TRAF3IP2, REL, and FBXL19, have been identified as having a significant impact on disease development. Interestingly, different genetic variants at the same locus can predispose individuals to either PsO or PsA (e.g., IL23R and deletion of LCE3B and LCE3C), with some variants being uniquely linked to PsA (like HLA B27 on chromosome 6). This article aims to summarize known and new data on the genetics of PsO and PsA, their associated genes, and the involvement of the HLA system and cytokines.
Collapse
Affiliation(s)
- Marek Zalesak
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia (L.D.)
| | - Lubos Danisovic
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia (L.D.)
- National Institute of Rheumatic Diseases, Nábrežie Ivana Krasku 4, 921 12 Piestany, Slovakia
| | - Stefan Harsanyi
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia (L.D.)
- National Institute of Rheumatic Diseases, Nábrežie Ivana Krasku 4, 921 12 Piestany, Slovakia
| |
Collapse
|
16
|
Liadaki K, Zafiriou E, Giannoulis T, Alexouda S, Chaidaki K, Gidarokosta P, Roussaki-Schulze AV, Tsiogkas SG, Daponte A, Mamuris Z, Bogdanos DP, Moschonas NK, Sarafidou T. PDE4 Gene Family Variants Are Associated with Response to Apremilast Treatment in Psoriasis. Genes (Basel) 2024; 15:369. [PMID: 38540428 PMCID: PMC10970167 DOI: 10.3390/genes15030369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/07/2024] [Accepted: 03/14/2024] [Indexed: 06/14/2024] Open
Abstract
Moderate-to-severe psoriasis (Ps) treatment includes systemic drugs and biological agents. Apremilast, a small molecule primarily metabolized by cytochrome CYP3A4, modulates the immune system by specifically inhibiting phosphodiesterase type 4 (PDE4) isoforms and is currently used for the treatment of Ps and psoriatic arthritis (PsA). Clinical trials and real-world data showed variable efficacy in response among Ps patients underlying the need for personalized therapy. This study implements a candidate-gene and a network-based approach to identify genetic markers associated with apremilast response in forty-nine Greek Ps patients. Our data revealed an association of sixty-four SNPs within or near PDE4 and CYP3A4 genes, four SNPs in ncRNAs ANRIL, LINC00941 and miR4706, which influence the abundance or function of PDE4s, and thirty-three SNPs within fourteen genes whose protein products either interact directly with PDE4 proteins or constitute components of the cAMP signaling pathway which is modulated by PDE4s. Notably, fifty-six of the aforementioned SNPs constitute eQTLs for the respective genes in relevant to psoriasis tissues/cells implying that these variants could be causal. Our analysis provides a number of novel genetic variants that, upon validation in larger cohorts, could be utilized as predictive markers regarding the response of Ps patients to apremilast treatment.
Collapse
Affiliation(s)
- Kalliopi Liadaki
- Department of Biochemistry and Biotechnology, University of Thessaly, Viopolis, 41500 Larissa, Greece; (K.L.); (Z.M.)
| | - Efterpi Zafiriou
- Department of Dermatology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Viopolis, 41500 Larissa, Greece; (E.Z.); (K.C.); (P.G.); (A.-V.R.-S.)
| | | | - Sofia Alexouda
- Department of Biochemistry and Biotechnology, University of Thessaly, Viopolis, 41500 Larissa, Greece; (K.L.); (Z.M.)
| | - Kleoniki Chaidaki
- Department of Dermatology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Viopolis, 41500 Larissa, Greece; (E.Z.); (K.C.); (P.G.); (A.-V.R.-S.)
| | - Polyxeni Gidarokosta
- Department of Dermatology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Viopolis, 41500 Larissa, Greece; (E.Z.); (K.C.); (P.G.); (A.-V.R.-S.)
| | - Angeliki-Viktoria Roussaki-Schulze
- Department of Dermatology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Viopolis, 41500 Larissa, Greece; (E.Z.); (K.C.); (P.G.); (A.-V.R.-S.)
| | - Sotirios G. Tsiogkas
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Viopolis, 41500 Larissa, Greece; (S.G.T.); (A.D.); (D.P.B.)
| | - Athina Daponte
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Viopolis, 41500 Larissa, Greece; (S.G.T.); (A.D.); (D.P.B.)
| | - Zissis Mamuris
- Department of Biochemistry and Biotechnology, University of Thessaly, Viopolis, 41500 Larissa, Greece; (K.L.); (Z.M.)
| | - Dimitrios P. Bogdanos
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Viopolis, 41500 Larissa, Greece; (S.G.T.); (A.D.); (D.P.B.)
| | - Nicholas K. Moschonas
- School of Medicine, University of Patras, 26500 Patras, Greece
- Foundation for Research and Technology Hellas, Institute of Chemical Engineering Sciences, 26504 Patras, Greece
| | - Theologia Sarafidou
- Department of Biochemistry and Biotechnology, University of Thessaly, Viopolis, 41500 Larissa, Greece; (K.L.); (Z.M.)
| |
Collapse
|
17
|
Coto-Segura P, Vázquez-Coto D, Velázquez-Cuervo L, García-Lago C, Coto E, Queiro R. The IFIH1/ MDA5 rs1990760 Gene Variant (946Thr) Differentiates Early- vs. Late-Onset Skin Disease and Increases the Risk of Arthritis in a Spanish Cohort of Psoriasis. Int J Mol Sci 2023; 24:14803. [PMID: 37834254 PMCID: PMC10572774 DOI: 10.3390/ijms241914803] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 08/29/2023] [Accepted: 09/29/2023] [Indexed: 10/15/2023] Open
Abstract
The melanoma differentiation-associated protein 5 (MDA5; encoded by the IFIH1 gene) mediates the activation of the interferon pathway in response to a viral infection. This protein is also upregulated in autoimmune diseases and psoriasis skin lesions. IFIH1 gene variants that increase MDA5 activity have been associated with an increased risk for immune-mediated diseases, including psoriasis. Our aim is to determine the association between three IFIH1 variants (rs35337543 G/C, intron8 + 1; rs35744605 C/A, Glu627Stop; and rs1990760 C/T, Ala946Thr) and the main clinical findings in a cohort of Spanish patients with psoriasis (N = 572; 77% early-onset). Early-onset psoriasis patients (EOPs) had a significantly higher frequency of severe disease and the Cw6*0602 allele. Carriers of rs1990760 T (946Thr) were more common in the EOPs (p < 0.001), and the effect was more pronounced among Cw6*0602-negatives. This variant was also associated with an increased risk of psoriatic arthritis (PsA) independent from other factors (OR = 1.62, 95%CI = 1.11-2.37). The rs3533754 and rs35744605 polymorphisms did not show significant differences between the two onset age or PsA groups. Compared to the controls, the 946Thr variant was more common in the EOPs (nonsignificant difference) and significantly less common in patients aged >40 years (p = 0.005). In conclusion, the common IFIH1 rs1990760 T allele was significantly more frequent in early-onset compared to late-onset patients. This variant was also an independent risk factor for PsA in our cohort. Our study reinforces the widely reported role of the IFIH1 gene variants on psoriatic disease.
Collapse
Affiliation(s)
- Pablo Coto-Segura
- Dermatología, Hospital Universitario Vital Alvarez-Buylla, 33011 Mieres, Spain;
| | - Daniel Vázquez-Coto
- Genética Molecular, Hospital Universitario Central Asturias, 33011 Oviedo, Spain; (D.V.-C.); (L.V.-C.); (C.G.-L.); (E.C.)
| | - Lucinda Velázquez-Cuervo
- Genética Molecular, Hospital Universitario Central Asturias, 33011 Oviedo, Spain; (D.V.-C.); (L.V.-C.); (C.G.-L.); (E.C.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Claudia García-Lago
- Genética Molecular, Hospital Universitario Central Asturias, 33011 Oviedo, Spain; (D.V.-C.); (L.V.-C.); (C.G.-L.); (E.C.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Eliecer Coto
- Genética Molecular, Hospital Universitario Central Asturias, 33011 Oviedo, Spain; (D.V.-C.); (L.V.-C.); (C.G.-L.); (E.C.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Departamento Medicina, Universidad de Oviedo, 33011 Oviedo, Spain
| | - Rubén Queiro
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Departamento Medicina, Universidad de Oviedo, 33011 Oviedo, Spain
- Reumatología, Hospital Universitario Central Asturias, 33011 Oviedo, Spain
| |
Collapse
|
18
|
Soomro M, Hum R, Barton A, Bowes J. Genetic Studies Investigating Susceptibility to Psoriatic Arthritis: A Narrative Review. Clin Ther 2023; 45:810-815. [PMID: 37516563 DOI: 10.1016/j.clinthera.2023.07.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 07/07/2023] [Accepted: 07/10/2023] [Indexed: 07/31/2023]
Abstract
PURPOSE Approximately 30% of patients with psoriasis will develop psoriatic arthritis (PsA), leading to a decreased quality of life for the patient caused by increasing disability and additional health complications. The identification of risk factors for the development of PsA would facilitate the development of risk prediction models in which patients with psoriasis at high risk of developing PsA could be targeted in a stratified medicine approach, enabling early intervention and treatment. PsA is known to have a genetic contribution to susceptibility, and the identification of genetic risk factors that differentiate PsA from cutaneous-only psoriasis is a key area of research. This narrative review summarizes the discovery of genetic risk factors and, with the aid of a primer on risk prediction models, discusses their potential role for the classification of PsA risk and diagnosis. METHODS All relevant research articles were identified through searches of the PubMed database for literature published up until December 2022. Search terms included psoriatic arthritis, genetic susceptibility, genetic association, genome-wide association study, GWAS, prediction, and polygenic risk score. FINDINGS The current literature reveals considerable overlap between the genetic susceptibility loci for PsA and psoriasis. Several PsA-specific genetic risk factors have been reported, and most notably these implicate the HLA-B and IL23R genes. Efforts to include genetic risk factors in prediction models for the development of PsA have reported good discrimination. IMPLICATIONS Key messages emerging from this narrative are as follows: the limited number of PsA-specific susceptibility loci reported to date suggest larger studies are required, facilitated by international collaboration, to achieve the power to detect further genetic factors; the early promising results for genetic-based risk prediction require further validation in independent datasets; and risk prediction models combining clinical and genetic risk factors have yet to be explored.
Collapse
Affiliation(s)
- Mehreen Soomro
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
| | - Ryan Hum
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
| | - Anne Barton
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom; NIHR Manchester Biomedical Research Centre, Central Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - John Bowes
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom; NIHR Manchester Biomedical Research Centre, Central Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom.
| |
Collapse
|
19
|
Navarro-Compán V, Puig L, Vidal S, Ramírez J, Llamas-Velasco M, Fernández-Carballido C, Almodóvar R, Pinto JA, Galíndez-Aguirregoikoa E, Zarco P, Joven B, Gratacós J, Juanola X, Blanco R, Arias-Santiago S, Sanz Sanz J, Queiro R, Cañete JD. The paradigm of IL-23-independent production of IL-17F and IL-17A and their role in chronic inflammatory diseases. Front Immunol 2023; 14:1191782. [PMID: 37600764 PMCID: PMC10437113 DOI: 10.3389/fimmu.2023.1191782] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 07/05/2023] [Indexed: 08/22/2023] Open
Abstract
Interleukin-17 family (IL-17s) comprises six structurally related members (IL-17A to IL-17F); sequence homology is highest between IL-17A and IL-17F, displaying certain overlapping functions. In general, IL-17A and IL-17F play important roles in chronic inflammation and autoimmunity, controlling bacterial and fungal infections, and signaling mainly through activation of the nuclear factor-kappa B (NF-κB) pathway. The role of IL-17A and IL-17F has been established in chronic immune-mediated inflammatory diseases (IMIDs), such as psoriasis (PsO), psoriatic arthritis (PsA), axial spondylarthritis (axSpA), hidradenitis suppurativa (HS), inflammatory bowel disease (IBD), multiple sclerosis (MS), and asthma. CD4+ helper T cells (Th17) activated by IL-23 are well-studied sources of IL-17A and IL-17F. However, other cellular subtypes can also produce IL-17A and IL-17F, including gamma delta (γδ) T cells, alpha beta (αβ) T cells, type 3 innate lymphoid cells (ILC3), natural killer T cells (NKT), or mucosal associated invariant T cells (MAIT). Interestingly, the production of IL-17A and IL-17F by innate and innate-like lymphocytes can take place in an IL-23 independent manner in addition to IL-23 classical pathway. This would explain the limitations of the inhibition of IL-23 in the treatment of patients with certain rheumatic immune-mediated conditions such as axSpA. Despite their coincident functions, IL-17A and IL-17F contribute independently to chronic tissue inflammation having somehow non-redundant roles. Although IL-17A has been more widely studied, both IL-17A and IL-17F are overexpressed in PsO, PsA, axSpA and HS. Therefore, dual inhibition of IL-17A and IL-17F could provide better outcomes than IL-23 or IL-17A blockade.
Collapse
Affiliation(s)
| | - Luis Puig
- Department of Dermatology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Silvia Vidal
- Immunology-Inflammatory Diseases, Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Julio Ramírez
- Arthritis Unit, Department of Rheumatology, Hospital Clínic and Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Mar Llamas-Velasco
- Department of Dermatology, Hospital Universitario La Princesa, Madrid, Spain
| | | | - Raquel Almodóvar
- Department of Rheumatology, Hospital Universitario Fundación Alcorcón, Alcorcón, Madrid, Spain
| | - José Antonio Pinto
- Department of Rheumatology, Complejo Hospitalario Universitario de A Coruña, Instituto de Investigación Biomédica de A Coruña (INIBIC), A Coruña, Spain
| | | | - Pedro Zarco
- Department of Rheumatology, Hospital Universitario Fundación Alcorcón, Alcorcón, Madrid, Spain
| | - Beatriz Joven
- Department of Rheumatology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Jordi Gratacós
- Department of Rheumatology, Medicine Department Autonomus University of Barcelona (UAB), I3PT, University Hospital Parc Taulí Sabadell, Barcelona, Spain
| | - Xavier Juanola
- Department of Rheumatology, University Hospital Bellvitge, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Ricardo Blanco
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - Salvador Arias-Santiago
- Department of Dermatology, Hospital Universitario Virgen de las Nieves, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
- Department of Dermatology, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Jesús Sanz Sanz
- Department of Rheumatology, Hospital Universitario Puerta del Hierro Majadahonda, Madrid, Spain
| | - Rubén Queiro
- Department of Rheumatology, Hospital Universitario Central de Asturias, Oviedo, Asturias, Spain
| | - Juan D. Cañete
- Arthritis Unit, Department of Rheumatology, Hospital Clínic and Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
20
|
Scrivo R, D'Angelo S, Carriero A, Castellani C, Perrotta FM, Conti F, Vecellio M, Selmi C, Lubrano E. The Conundrum of Psoriatic Arthritis: a Pathogenetic and Clinical Pattern at the Midpoint of Autoinflammation and Autoimmunity. Clin Rev Allergy Immunol 2023; 65:72-85. [PMID: 35040085 DOI: 10.1007/s12016-021-08914-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/20/2021] [Indexed: 02/06/2023]
Abstract
Psoriatic arthritis (PsA) is a chronic inflammatory condition characterized by psoriasis, synovitis, enthesitis, spondylitis, and the possible association with other extra-articular manifestations and comorbidities. It is a multifaceted and systemic disorder sustained by complex pathogenesis, combining aspects of autoinflammation and autoimmunity. Features of PsA autoinflammation include the role of biomechanical stress in the onset and/or exacerbation of the disease; the evidence of involvement of the innate immune response mediators in the skin, peripheral blood and synovial tissue; an equal gender distribution; the clinical course which may encounter periods of prolonged remission and overlapping features with autoinflammatory syndromes. Conversely, the role of autoimmunity is evoked by the association with class I major histocompatibility complex alleles, the polyarticular pattern of the disease which sometimes resembles rheumatoid arthritis and the presence of serum autoantibodies. Genetics also provide important insights into the pathogenesis of PsA, particularly related to class I HLA being associated with psoriasis and PsA. In this review, we provide a comprehensive review of the pathogenesis, genetics and clinical features of PsA that endorse the mixed nature of a disorder at the crossroads of autoinflammation and autoimmunity.
Collapse
Affiliation(s)
- Rossana Scrivo
- Rheumatology Unit, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Salvatore D'Angelo
- Rheumatology Institute of Lucania (IReL) - Rheumatology Department of Lucania, San Carlo Hospital of Potenza and Madonna Delle Grazie Hospital of Matera, Potenza, Italy
| | - Antonio Carriero
- Rheumatology Institute of Lucania (IReL) - Rheumatology Department of Lucania, San Carlo Hospital of Potenza and Madonna Delle Grazie Hospital of Matera, Potenza, Italy
- Dipartimento Di Medicina E Scienze Della Salute, Università Degli Studi del Molise, Campobasso, Italy
| | - Chiara Castellani
- Rheumatology Unit, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Fabio Massimo Perrotta
- Dipartimento Di Medicina E Scienze Della Salute, Università Degli Studi del Molise, Campobasso, Italy
| | - Fabrizio Conti
- Rheumatology Unit, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Matteo Vecellio
- Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center - IRCCS, Rozzano, MI, Italy
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Botnar Research Center, Oxford, UK
| | - Carlo Selmi
- Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center - IRCCS, Rozzano, MI, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, MI, Italy
| | - Ennio Lubrano
- Dipartimento Di Medicina E Scienze Della Salute, Università Degli Studi del Molise, Campobasso, Italy
| |
Collapse
|
21
|
Patrick MT, Nair RP, He K, Stuart PE, Billi AC, Zhou X, Gudjonsson JE, Oksenberg JR, Elder JT, Tsoi LC. Shared Genetic Risk Factors for Multiple Sclerosis/Psoriasis Suggest Involvement of Interleukin-17 and Janus Kinase-Signal Transducers and Activators of Transcription Signaling. Ann Neurol 2023; 94:384-397. [PMID: 37127916 PMCID: PMC10524664 DOI: 10.1002/ana.26672] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 04/24/2023] [Accepted: 04/27/2023] [Indexed: 05/03/2023]
Abstract
OBJECTIVE Psoriasis and multiple sclerosis (MS) are complex immune diseases that are mediated by T cells and share multiple comorbidities. Previous studies have suggested psoriatic patients are at higher risk of MS; however, causal relationships between the two conditions remain unclear. Through epidemiology and genetics, we provide a comprehensive understanding of the relationship, and share molecular factors between psoriasis and MS. METHODS We used logistic regression, trans-disease meta-analysis and Mendelian randomization. Medical claims data were included from 30 million patients, including 141,544 with MS and 742,919 with psoriasis. We used genome-wide association study summary statistics from 11,024 psoriatic, 14,802 MS cases, and 43,039 controls for trans-disease meta-analysis, with additional summary statistics from 5 million individuals for Mendelian randomization. RESULTS Psoriatic patients have a significantly higher risk of MS (4,637 patients with both diseases; odds ratio [OR] 1.07, p = 1.2 × 10-5 ) after controlling for potential confounders. Using inverse variance and equally weighted trans-disease meta-analysis, we revealed >20 shared and opposing (direction of effect) genetic loci outside the major histocompatibility complex that showed significant genetic colocalization (in COLOC and COLOC-SuSiE v5.1.0). Co-expression analysis of genes from these loci further identified distinct clusters that were enriched among pathways for interleukin-17/tumor necrosis factor-α (OR >39, p < 1.6 × 10-3 ) and Janus kinase-signal transducers and activators of transcription (OR 35, p = 1.1 × 10-5 ), including genes, such as TNFAIP3, TYK2, and TNFRSF1A. Mendelian randomization found psoriasis as an exposure has a significant causal effect on MS (OR 1.04, p = 5.8 × 10-3 ), independent of type 1 diabetes (OR 1.05, p = 4.3 × 10-7 ), type 2 diabetes (OR 1.08, p = 2.3 × 10-3 ), inflammatory bowel disease (OR 1.11, p = 1.6 × 10-11 ), and vitamin D level (OR 0.75, p = 9.4 × 10-3 ). INTERPRETATION By investigating the shared genetics of psoriasis and MS, along with their modifiable risk factors, our findings will advance innovations in treatment for patients suffering from comorbidities. ANN NEUROL 2023;94:384-397.
Collapse
Affiliation(s)
- Matthew T. Patrick
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Rajan P. Nair
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Kevin He
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Philip E. Stuart
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Allison C. Billi
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Xiang Zhou
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Johann E. Gudjonsson
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Jorge R. Oksenberg
- Department of Neurology, University of California, San Francisco, California, United States of America
| | - James T. Elder
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Lam C. Tsoi
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
22
|
Sánchez-Rodríguez G, Puig L. Pathogenic Role of IL-17 and Therapeutic Targeting of IL-17F in Psoriatic Arthritis and Spondyloarthropathies. Int J Mol Sci 2023; 24:10305. [PMID: 37373452 DOI: 10.3390/ijms241210305] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/08/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
The interleukin 17 (IL-17) family, a subset of cytokines consisting of IL-17A-F, plays crucial roles in host defence against microbial organisms and the development of inflammatory diseases, including psoriasis (PsO), axial spondyloarthritis (axSpA), and psoriatic arthritis (PsA). IL-17A is the signature cytokine produced by T helper 17 (Th17) cells and is considered the most biologically active form. The pathogenetic involvement of IL-17A in these conditions has been confirmed, and its blockade with biological agents has provided a highly effective therapeutical approach. IL-17F is also overexpressed in the skin and synovial tissues of patients with these diseases, and recent studies suggest its involvement in promoting inflammation and tissue damage in axSpA and PsA. The simultaneous targeting of IL-17A and IL-17F by dual inhibitors and bispecific antibodies may improve the management of Pso, PsA, and axSpA, as demonstrated in the pivotal studies of dual specific antibodies such as bimekizumab. The present review focuses on the role of IL-17F and its therapeutic blockade in axSpA and PsA.
Collapse
Affiliation(s)
- Guillermo Sánchez-Rodríguez
- Department of Dermatology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Carrer de Sant Quintí, 89, 08041 Barcelona, Spain
| | - Lluís Puig
- Department of Dermatology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Carrer de Sant Quintí, 89, 08041 Barcelona, Spain
| |
Collapse
|
23
|
Kuiper JJ, Prinz JC, Stratikos E, Kuśnierczyk P, Arakawa A, Springer S, Mintoff D, Padjen I, Shumnalieva R, Vural S, Kötter I, van de Sande MG, Boyvat A, de Boer JH, Bertsias G, de Vries N, Krieckaert CL, Leal I, Vidovič Valentinčič N, Tugal-Tutkun I, El Khaldi Ahanach H, Costantino F, Glatigny S, Mrazovac Zimak D, Lötscher F, Kerstens FG, Bakula M, Viera Sousa E, Böhm P, Bosman K, Kenna TJ, Powis SJ, Breban M, Gul A, Bowes J, Lories RJ, Nowatzky J, Wolbink GJ, McGonagle DG, Turkstra F. EULAR study group on ‘MHC-I-opathy’: identifying disease-overarching mechanisms across disciplines and borders. Ann Rheum Dis 2023:ard-2022-222852. [PMID: 36987655 DOI: 10.1136/ard-2022-222852] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 01/25/2023] [Indexed: 03/29/2023]
Abstract
The ‘MHC-I (major histocompatibility complex class I)-opathy’ concept describes a family of inflammatory conditions with overlapping clinical manifestations and a strong genetic link to the MHC-I antigen presentation pathway. Classical MHC-I-opathies such as spondyloarthritis, Behçet’s disease, psoriasis and birdshot uveitis are widely recognised for their strong association with certain MHC-I alleles and gene variants of the antigen processing aminopeptidases ERAP1 and ERAP2 that implicates altered MHC-I peptide presentation to CD8+T cells in the pathogenesis. Progress in understanding the cause and treatment of these disorders is hampered by patient phenotypic heterogeneity and lack of systematic investigation of the MHC-I pathway.Here, we discuss new insights into the biology of MHC-I-opathies that strongly advocate for disease-overarching and integrated molecular and clinical investigation to decipher underlying disease mechanisms. Because this requires transformative multidisciplinary collaboration, we introduce the EULAR study group on MHC-I-opathies to unite clinical expertise in rheumatology, dermatology and ophthalmology, with fundamental and translational researchers from multiple disciplines such as immunology, genomics and proteomics, alongside patient partners. We prioritise standardisation of disease phenotypes and scientific nomenclature and propose interdisciplinary genetic and translational studies to exploit emerging therapeutic strategies to understand MHC-I-mediated disease mechanisms. These collaborative efforts are required to address outstanding questions in the etiopathogenesis of MHC-I-opathies towards improving patient treatment and prognostication.
Collapse
Affiliation(s)
- Jonas Jw Kuiper
- Department of Ophthalmology, Center for Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Jörg C Prinz
- University Hospital, department of Dermatology and Allergy, Ludwig Maximilians University Munich, Munchen, Germany
| | - Efstratios Stratikos
- Laboratory of Biochemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Piotr Kuśnierczyk
- Laboratory of Immunogenetics and Tissue Immunology, Institute of Immunology and Experimental Therapy Ludwik Hirszfeld Polish Academy of Sciences, Wroclaw, Poland
| | - Akiko Arakawa
- University Hospital, department of Dermatology and Allergy, Ludwig Maximilians University Munich, Munchen, Germany
| | | | - Dillon Mintoff
- Department of Dermatology, Mater Dei Hospital, Msida, Malta
- Department of Pathology, University of Malta Faculty of Medicine and Surgery, Msida, Malta
| | - Ivan Padjen
- Division of Clinical Immunology and Rheumatology, University Hospital Centre Zagreb Department of Internal Medicine, Zagreb, Croatia
- University of Zagreb School of Medicine, Zagreb, Croatia
| | - Russka Shumnalieva
- Clinic of Rheumatology, Department of Rheumatology, Medical University of Sofia, Sofia, Bulgaria
| | - Seçil Vural
- School of Medicine, Department of Dermatology, Koç University, Istanbul, Turkey
| | - Ina Kötter
- Clinic for Rheumatology and Immunology, Bad Bramdsted Hospital, Bad Bramstedt, Germany
- Division of Rheumatology and Systemic Inflammatory Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marleen G van de Sande
- University of Amsterdam, Department of Rheumatology & Clinical Immunology and Department of Experimental Immunology, Amsterdam Institute for Infection & Immunity, Amsterdam UMC Location AMC, Amsterdam, The Netherlands
- Amsterdam Rheumatology and Immunology Center (ARC) | Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ayşe Boyvat
- Department of Dermatology, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Joke H de Boer
- Department of Ophthalmology, Center for Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - George Bertsias
- Department of Rheumatology and Clinical Immunology, University of Crete School of Medicine, Iraklio, Greece
- Laboratory of Autoimmunity-Inflammation, Institute of Molecular Biology and Biotechnology, Heraklion, Greece
| | - Niek de Vries
- University of Amsterdam, Department of Rheumatology & Clinical Immunology and Department of Experimental Immunology, Amsterdam Institute for Infection & Immunity, Amsterdam UMC Location AMC, Amsterdam, The Netherlands
- Amsterdam Rheumatology and Immunology Center (ARC) | Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Charlotte Lm Krieckaert
- Amsterdam Rheumatology and immunology Center (ARC)| Reade, Amsterdam, The Netherlands
- Department of Rheumatology, Reade Hoofdlocatie Dr Jan van Breemenstraat, Amsterdam, The Netherlands
| | - Inês Leal
- Department of Ophthalmology, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte EPE, Lisboa, Portugal
- Centro de Estudeos das Ciencias da Visão, Universidade de Lisboa Faculdade de Medicina, Lisboa, Portugal
| | - Nataša Vidovič Valentinčič
- University Eye Clinic, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Ilknur Tugal-Tutkun
- Department of Ophthalmology, Istanbul University Istanbul Faculty of Medicine, Istanbul, Turkey
| | - Hanane El Khaldi Ahanach
- Departement of Ophthalmology, Amsterdam UMC Location AMC, Amsterdam, The Netherlands
- Department of Ophthalmology, Onze Lieve Vrouwe Gasthuis, Amsterdam, The Netherlands
| | - Félicie Costantino
- Service de Rheumatology, Hospital Ambroise-Pare, Boulogne-Billancourt, France
- Infection & Inflammation, UMR 1173, Inserm, UVSQ, University Paris-Saclay, Montigny-le-Bretonneux, France
| | - Simon Glatigny
- Infection & Inflammation, UMR 1173, Inserm, UVSQ/Université Paris Saclay, Montigny-le-Bretonneux, France
- Laboratoire d'Excellence Inflamex, Paris, France
| | | | - Fabian Lötscher
- Department of Rheumatology and Immunology, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland
| | - Floor G Kerstens
- Amsterdam Rheumatology and immunology Center (ARC)| Reade, Amsterdam, The Netherlands
- Department of Rheumatology, Reade Hoofdlocatie Dr Jan van Breemenstraat, Amsterdam, The Netherlands
| | - Marija Bakula
- Division of Clinical Immunology and Rheumatology, University Hospital Centre Zagreb Department of Internal Medicine, Zagreb, Croatia
| | - Elsa Viera Sousa
- Rheumatology Research Unit Molecular João Lobo Antunes, University of Lisbon Medical Faculty, Lisboa, Portugal
- Rheumatology DepartmentSanta Maria Centro Hospital, Academic Medical Centre of Lisbon, Lisboa, Portugal
| | - Peter Böhm
- Patientpartner, German League against Rheumatism, Bonn, Germany
| | - Kees Bosman
- Patientpartner, Nationale Vereniging ReumaZorg, Nijmegen, The Netherlands
| | - Tony J Kenna
- Translational Research Institute, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Simon J Powis
- School of Medicine, University of St Andrews School of Medicine, St Andrews, UK
| | - Maxime Breban
- Service de Rheumatology, Hospital Ambroise-Pare, Boulogne-Billancourt, France
- Infection & Inflammation, UMR 1173, Inserm, UVSQ, University Paris-Saclay, Montigny-le-Bretonneux, France
| | - Ahmet Gul
- Division of Rheumatology, Istanbul University Istanbul Faculty of Medicine, Istanbul, Turkey
| | - John Bowes
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Center, The University of Manchester, Manchester, UK
- NIHR Manchester Musculoskeletal Biomedical Research Unit, Manchester University NHS Foundation Trust, Manchester, UK
| | - Rik Ju Lories
- Department of Rheumatology, KU Leuven University Hospitals Leuven, Leuven, Belgium
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Johannes Nowatzky
- Department of Medicine, Division of Rheumatology, NYU Langone Behçet's Disease Program, NYU Langone Ocular Rheumatology Program, New York University Grossman School of Medicine, New York University, New York, New York, USA
- Department of Pathology, New York University Grossman School of Medicine, New York, New York, USA
| | - Gerrit Jan Wolbink
- Amsterdam Rheumatology and immunology Center (ARC)| Reade, Amsterdam, The Netherlands
- Department Immunopathology, Sanquin Research, Amsterdam, The Netherlands
| | - Dennis G McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Franktien Turkstra
- Amsterdam Rheumatology and immunology Center (ARC)| Reade, Amsterdam, The Netherlands
- Department of Rheumatology, Reade Hoofdlocatie Dr Jan van Breemenstraat, Amsterdam, The Netherlands
| |
Collapse
|
24
|
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: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [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
|
25
|
Targeting the transcription factor HES1 by L-menthol restores protein phosphatase 6 in keratinocytes in models of psoriasis. Nat Commun 2022; 13:7815. [PMID: 36535970 PMCID: PMC9763329 DOI: 10.1038/s41467-022-35565-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Protein Phosphatase 6 down-regulation in keratinocytes is a pivotal event that amplifies the inflammatory circuits in psoriasis, indicating that restoration of protein phosphatase 6 can be a rational strategy for psoriasis treatment. Through the phenotypic screen, we here identify L-menthol that ameliorates psoriasis-like skin inflammation by increasing protein phosphatase 6 in keratinocytes. Target identification approaches reveal an indispensable role for the transcription factor hairy and enhancer of split 1 in governing the protein phosphatase 6-upregulating function of L-menthol in keratinocytes. The transcription factor hairy and enhancer of split 1 is diminished in the epidermis of psoriasis patients and imiquimod-induced mouse model, while L-menthol upregulates the transcription factor hairy and enhancer of split 1 by preventing its proteasomal degradation. Mechanistically, the transcription factor hairy and enhancer of split 1 transcriptionally activates the expression of immunoglobulin-binding protein 1 which promotes protein phosphatase 6 expression and inhibits its ubiquitination. Collectively, we discover a therapeutic compound, L-menthol, for psoriasis, and uncover the dysfunctional the transcription factor hairy and enhancer of split 1- immunoglobulin-binding protein 1- protein phosphatase 6 axis that contributes to psoriasis pathology by using L-menthol as a probe.
Collapse
|
26
|
Packham J, Tarar B. An overview of psoriatic arthritis including clinical manifestations, assessment, diagnostic criteria, investigations, drug management and GRAPPA guidelines. Musculoskeletal Care 2022; 20 Suppl 1:S2-S11. [PMID: 36356108 DOI: 10.1002/msc.1693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 08/15/2022] [Indexed: 06/16/2023]
Abstract
Psoriatic arthritis (PsA) is a chronic and often progressive inflammatory disease, occurring in up to 30% of patients with psoriasis. Assessment of patients with PsA requires consideration of all disease domains, including peripheral arthritis, axial disease, enthesitis, dactylitis, skin psoriasis, psoriatic nail disease, uveitis and inflammatory bowel disease. Co-morbidities and related conditions should all be considered including: obesity, metabolic syndrome, cardiovascular disease, anxiety/depression, liver disease, chronic infections, malignancy, osteoporosis, fibromyalgia and reproductive health.
Collapse
Affiliation(s)
- Jonathan Packham
- Haywood Rheumatology Centre, Midlands Partnership NHS Foundation Trust, Staffordshire, UK
- Academic Unit of Population and Lifespan Sciences, University of Nottingham, Nottingham, UK
| | - Bilal Tarar
- Haywood Rheumatology Centre, Midlands Partnership NHS Foundation Trust, Staffordshire, UK
| |
Collapse
|
27
|
Gurke R, Bendes A, Bowes J, Koehm M, Twyman RM, Barton A, Elewaut D, Goodyear C, Hahnefeld L, Hillenbrand R, Hunter E, Ibberson M, Ioannidis V, Kugler S, Lories RJ, Resch E, Rüping S, Scholich K, Schwenk JM, Waddington JC, Whitfield P, Geisslinger G, FitzGerald O, Behrens F, Pennington SR, on behalf of the HIPPOCRATES Consortium. Omics and Multi-Omics Analysis for the Early Identification and Improved Outcome of Patients with Psoriatic Arthritis. Biomedicines 2022; 10:2387. [PMID: 36289648 PMCID: PMC9598654 DOI: 10.3390/biomedicines10102387] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/15/2022] [Accepted: 09/17/2022] [Indexed: 11/17/2022] Open
Abstract
The definitive diagnosis and early treatment of many immune-mediated inflammatory diseases (IMIDs) is hindered by variable and overlapping clinical manifestations. Psoriatic arthritis (PsA), which develops in ~30% of people with psoriasis, is a key example. This mixed-pattern IMID is apparent in entheseal and synovial musculoskeletal structures, but a definitive diagnosis often can only be made by clinical experts or when an extensive progressive disease state is apparent. As with other IMIDs, the detection of multimodal molecular biomarkers offers some hope for the early diagnosis of PsA and the initiation of effective management and treatment strategies. However, specific biomarkers are not yet available for PsA. The assessment of new markers by genomic and epigenomic profiling, or the analysis of blood and synovial fluid/tissue samples using proteomics, metabolomics and lipidomics, provides hope that complex molecular biomarker profiles could be developed to diagnose PsA. Importantly, the integration of these markers with high-throughput histology, imaging and standardized clinical assessment data provides an important opportunity to develop molecular profiles that could improve the diagnosis of PsA, predict its occurrence in cohorts of individuals with psoriasis, differentiate PsA from other IMIDs, and improve therapeutic responses. In this review, we consider the technologies that are currently deployed in the EU IMI2 project HIPPOCRATES to define biomarker profiles specific for PsA and discuss the advantages of combining multi-omics data to improve the outcome of PsA patients.
Collapse
Affiliation(s)
- Robert Gurke
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Annika Bendes
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 171 65 Solna, Sweden
| | - John Bowes
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9WU, UK
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester M13 9PT, UK
| | - Michaela Koehm
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Division of Rheumatology, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | | | - Anne Barton
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9WU, UK
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester M13 9PT, UK
| | - Dirk Elewaut
- VIB-UGent Center for Inflammation Research, Ghent University, 9052 Ghent, Belgium
| | - Carl Goodyear
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8QQ, UK
| | - Lisa Hahnefeld
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | | | - Ewan Hunter
- Oxford BioDynamics Limited, Oxford OX4 2JZ, UK
| | - Mark Ibberson
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland
| | - Vassilios Ioannidis
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland
| | - Sabine Kugler
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer IAIS, Institute for Intelligent Analysis and Information Systems, Schloss Birlinghoven 1, 53757 Sankt Augustin, Germany
| | - Rik J. Lories
- Department of Development and Regeneration, KU Leuven, Skeletal Biology and Engineering Research Centre, P.O. Box 813 O&N, Herestraat 49, 3000 Leuven, Belgium
| | - Eduard Resch
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Stefan Rüping
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer IAIS, Institute for Intelligent Analysis and Information Systems, Schloss Birlinghoven 1, 53757 Sankt Augustin, Germany
| | - Klaus Scholich
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Jochen M. Schwenk
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 171 65 Solna, Sweden
| | - James C. Waddington
- Atturos Ltd., c/o UCD Conway Institute, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Phil Whitfield
- Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, Garscube Campus, University of Glasgow, Glasgow G61 1QH, UK
| | - Gerd Geisslinger
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Oliver FitzGerald
- UCD Conway Institute, School of Medicine, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Frank Behrens
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Division of Rheumatology, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Stephen R. Pennington
- Atturos Ltd., c/o UCD Conway Institute, University College Dublin, D04 V1W8 Dublin, Ireland
- UCD Conway Institute, School of Medicine, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | | |
Collapse
|
28
|
Atschekzei F, Dubrowinskaja N, Anim M, Thiele T, Witte T, Sogkas G. Identification of variants in genes associated with autoinflammatory disorders in a cohort of patients with psoriatic arthritis. RMD Open 2022; 8:rmdopen-2022-002561. [PMID: 36113963 PMCID: PMC9486391 DOI: 10.1136/rmdopen-2022-002561] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/21/2022] [Indexed: 11/18/2022] Open
Abstract
Objectives Besides adaptive immunity genes, genetic risk factors for psoriatic arthritis (PsA) include innate immunity loci, which suggests an autoinflammatory disease mechanism, at least in a subset of patients. Here, we aimed at investigating the autoinflammatory genetic background of PsA. Methods A total of 120 patients with PsA visiting the outpatient clinics of the Hannover University hospital underwent targeted next-generation sequencing, searching for variations in genes linked with inborn errors of immunity classified as autoinflammatory disorders (AIDs). Deleteriousness of rare variants was evaluated through in silico analysis. Results We found 45 rare predicted deleterious variants in 37 out of 120 (30.8%) patients with PsA. Relatively common were variants in AP1S3, PLCG2, NOD2 and NLRP12. All 45 variants were monoallelic and 25 of them, identified in 20 out of 120 (16.7%) patients, were localised in genes associated with autosomal dominant (AD) disorders. Detection of those variants is associated with pustular psoriasis or a coexisting inflammatory bowel disease (IBD). Conclusions Approximately 30% of patients with PsA harboured at least one variant in a gene associated with an AID, suggesting an autoinflammatory disease mechanism. Detection of variants in genes linked to AD-AIDs may explain extra-articular manifestations of PsA, such as pustular psoriasis and IBD.
Collapse
Affiliation(s)
| | | | - Manfred Anim
- Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Thea Thiele
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Torsten Witte
- Clinical Immunology, Hannover Medical School, Hannover, Germany
| | - Georgios Sogkas
- Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
29
|
Gisondi P, Bellinato F, Maurelli M, Geat D, Zabotti A, McGonagle D, Girolomoni G. Reducing the Risk of Developing Psoriatic Arthritis in Patients with Psoriasis. PSORIASIS (AUCKLAND, N.Z.) 2022; 12:213-220. [PMID: 35975124 PMCID: PMC9375973 DOI: 10.2147/ptt.s323300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 08/04/2022] [Indexed: 01/22/2023]
Abstract
Psoriatic arthritis (PsA) is a heterogeneous chronic inflammatory arthritis associated with psoriasis, which may manifest with different domains such as dactylitis, enthesitis, synovitis and spondylitis. The estimated prevalence of PsA in patients with psoriasis ranges widely between 6% and 42%. In most cases, PsA is preceded by skin involvement by an average time of 7-8 years. In the complex patho-mechanisms involved in the transition from psoriasis to PsA, the gut and skin have been proposed as the sites of immune activation triggering or contributing to the development of PsA. In such a transition, a subclinical phase has been identified, characterized by enthesopathy where soluble biomarkers and imaging findings but no clinical symptoms are detectable. Recent studies have provided some evidence that timely treated psoriasis may reduce the risk of developing PsA.
Collapse
Affiliation(s)
- Paolo Gisondi
- Section of Dermatology and Venereology, Department of Medicine, University of Verona, Verona, Italy
| | - Francesco Bellinato
- Section of Dermatology and Venereology, Department of Medicine, University of Verona, Verona, Italy
| | - Martina Maurelli
- Section of Dermatology and Venereology, Department of Medicine, University of Verona, Verona, Italy
| | - Davide Geat
- Section of Dermatology and Venereology, Department of Medicine, University of Verona, Verona, Italy
| | - Alen Zabotti
- Rheumatology Clinic, Department of Medical and Biological Sciences, University Hospital ‘Santa Maria della Misericordia’ c/o University of Udine, Udine, Italy
| | - Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Giampiero Girolomoni
- Section of Dermatology and Venereology, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
30
|
Molecular and cellular regulation of psoriatic inflammation. Clin Sci (Lond) 2022; 136:935-952. [PMID: 35730381 DOI: 10.1042/cs20210916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 02/06/2023]
Abstract
This review highlights the molecular and cellular mechanisms underlying psoriatic inflammation with an emphasis on recent developments which may impact on treatment approaches for this chronic disease. We consider both the skin and the musculoskeletal compartment and how different manifestations of psoriatic inflammation are linked. This review brings a focus to the importance of inflammatory feedback loops that exist in the initiation and chronic stages of the condition, and how close interaction between the epidermis and both innate and adaptive immune compartments drives psoriatic inflammation. Furthermore, we highlight work done on biomarkers to predict the outcome of therapy as well as the transition from psoriasis to psoriatic arthritis.
Collapse
|
31
|
Atzeni F, Siragusano C, Masala IF, Antonio C, Valentina P, D'Angelo S. IL-23 in axial spondyloarthritis and psoriatic arthritis: a good fit for biological treatment? Expert Opin Biol Ther 2022; 22:843-853. [PMID: 35722768 DOI: 10.1080/14712598.2022.2090834] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Interleukin 23 (IL-23) is a pro-inflammatory cytokine that plays a protective role against bacterial and fungal infections. However, the dysregulation of the IL-23/IL-17 axis provides a solid substrate for the development of various inflammatory diseases, such as psoriatic arthritis (PsA) and ankylosing spondylitis (AS). AREAS COVERED In different clinical trials, several drugs against IL-23 have shown efficacy and safety towards PsA, with excellent results on skin and joint scores. However, the same drugs did not show the same efficacy in AS, suggesting that IL-23 may not be a relevant driver of the pathobiology and clinical symptoms of active axial spondyloarthritis (axSpA). EXPERT OPINION These drugs have shown an excellent efficacy and a good safety profile towards PsA, while in AS the efficacy of the IL-23 blockade is lacking for reasons not yet known. Several hypotheses have been reported, but further studies will be needed for a greater understanding. This suggests the involvement of pathways or mechanisms for the development of SpA that remain unknown. In order to allow a wide use of IL-23 inhibitors, further clinical trials and long-term prospective studies are necessary.
Collapse
Affiliation(s)
- Fabiola Atzeni
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Cesare Siragusano
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | | | - Carriero Antonio
- Rheumatology Institute of Lucania (IReL): Rheumatology Department of Lucania, San Carlo Hospital of Potenza and Madonna delle Grazie Hospital of Matera, Potenza, Italy.,PhD Scholarship in Translational and Clinical Medicine, Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| | - Picerno Valentina
- Rheumatology Institute of Lucania (IReL): Rheumatology Department of Lucania, San Carlo Hospital of Potenza and Madonna delle Grazie Hospital of Matera, Potenza, Italy
| | - Salvatore D'Angelo
- Rheumatology Institute of Lucania (IReL): Rheumatology Department of Lucania, San Carlo Hospital of Potenza and Madonna delle Grazie Hospital of Matera, Potenza, Italy
| |
Collapse
|
32
|
Schett G, Rahman P, Ritchlin C, McInnes IB, Elewaut D, Scher JU. Psoriatic arthritis from a mechanistic perspective. Nat Rev Rheumatol 2022; 18:311-325. [PMID: 35513599 DOI: 10.1038/s41584-022-00776-6] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2022] [Indexed: 12/13/2022]
Abstract
Psoriatic arthritis (PsA) is part of a group of closely related clinical phenotypes ('psoriatic disease') that is defined by shared molecular pathogenesis resulting in excessive, prolonged inflammation in the various tissues affected, such as the skin, the entheses or the joints. Psoriatic disease comprises a set of specific drivers that promote an aberrant immune response and the consequent development of chronic disease that necessitates therapeutic intervention. These drivers include genetic, biomechanical, metabolic and microbial factors that facilitate a robust and continuous mobilization, trafficking and homing of immune cells into the target tissues. The role of genetic variants involved in the immune response, the contribution of mechanical factors triggering an exaggerated inflammatory response (mechanoinflammation), the impact of adipose tissue and altered lipid metabolism and the influence of intestinal dysbiosis in the disease process are discussed. Furthermore, the role of key cytokines, such as IL-23, IL-17 and TNF, in orchestrating the various phases of the inflammatory disease process and as therapeutic targets in PsA is reviewed. Finally, the nature and the mechanisms of inflammatory tissue responses inherent to PsA are summarized.
Collapse
Affiliation(s)
- Georg Schett
- Department of Medicine 3, Friedrich Alexander University Erlangen-Nuremberg and Universitaets-klinikum Erlangen, Erlangen, Germany. .,Deutsches Zentrum Immuntherapie DZI, Friedrich Alexander University Erlangen-Nuremberg and Universitaets-klinikum Erlangen, Erlangen, Germany.
| | - Proton Rahman
- St. Clare's Mercy Hospital, St. John's, Newfoundland, Canada
| | - Christopher Ritchlin
- Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center Rochester, Rochester, NY, USA
| | - Iain B McInnes
- College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Dirk Elewaut
- VIB-UGent Center for Inflammation Research and Ghent University Hospital, Ghent, Belgium
| | - Jose U Scher
- Department of Medicine, Division of Rheumatology, NYU Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
33
|
Soomro M, Stadler M, Dand N, Bluett J, Jadon D, Jalali-Najafabadi F, Duckworth M, Ho P, Marzo-Ortega H, Helliwell PS, Ryan AW, Kane D, Korendowych E, Simpson MA, Packham J, McManus R, Gabay C, Lamacchia C, Nissen MJ, Brown MA, Verstappen SMM, Van Staa T, Barker JN, Smith CH, FitzGerald O, McHugh N, Warren RB, Bowes J, Barton A. Comparative genetic analysis of psoriatic arthritis and psoriasis for the discovery of genetic risk factors and risk prediction modelling. Arthritis Rheumatol 2022; 74:1535-1543. [PMID: 35507331 PMCID: PMC9539852 DOI: 10.1002/art.42154] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 03/16/2022] [Accepted: 04/28/2022] [Indexed: 11/10/2022]
Abstract
Objectives Psoriatic arthritis (PsA) has a strong genetic component, and the identification of genetic risk factors could help identify the ~30% of psoriasis patients at high risk of developing PsA. Our objectives were to identify genetic risk factors and pathways that differentiate PsA from cutaneous‐only psoriasis (PsC) and to evaluate the performance of PsA risk prediction models. Methods Genome‐wide meta‐analyses were conducted separately for 5,065 patients with PsA and 21,286 healthy controls and separately for 4,340 patients with PsA and 6,431 patients with PsC. The heritability of PsA was calculated as a single‐nucleotide polymorphism (SNP)–based heritability estimate (h2SNP) and biologic pathways that differentiate PsA from PsC were identified using Priority Index software. The generalizability of previously published PsA risk prediction pipelines was explored, and a risk prediction model was developed with external validation. Results We identified a novel genome‐wide significant susceptibility locus for the development of PsA on chromosome 22q11 (rs5754467; P = 1.61 × 10−9), and key pathways that differentiate PsA from PsC, including NF‐κB signaling (adjusted P = 1.4 × 10−45) and Wnt signaling (adjusted P = 9.5 × 10−58). The heritability of PsA in this cohort was found to be moderate (h2SNP = 0.63), which was similar to the heritability of PsC (h2SNP = 0.61). We observed modest performance of published classification pipelines (maximum area under the curve 0.61), with similar performance of a risk model derived using the current data. Conclusion Key biologic pathways associated with the development of PsA were identified, but the investigation of risk classification revealed modest utility in the available data sets, possibly because many of the PsC patients included in the present study were receiving treatments that are also effective in PsA. Future predictive models of PsA should be tested in PsC patients recruited from primary care.
Collapse
Affiliation(s)
- Mehreen Soomro
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester, UK
| | - Michael Stadler
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester, UK
| | - Nick Dand
- Department of Medical and Molecular Genetics, School of Basic & Medical Biosciences, King's College London, London, UK
| | - James Bluett
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester, UK.,NIHR Manchester Musculoskeletal Biomedical Research Unit, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, UK
| | - Deepak Jadon
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Farideh Jalali-Najafabadi
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester, UK
| | - Michael Duckworth
- St John's Institute of Dermatology, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Pauline Ho
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester, UK.,NIHR Manchester Musculoskeletal Biomedical Research Unit, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, UK
| | - Helena Marzo-Ortega
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals Trust and Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, UK
| | - Philip S Helliwell
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals Trust and Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, UK
| | - Anthony W Ryan
- Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Ireland.,Genuity Science, Cherrywood Business Park, Dublin, Ireland
| | - David Kane
- Tallaght University Hospital and Trinity College Dublin, Ireland
| | - Eleanor Korendowych
- Royal National Hospital for Rheumatic Diseases and Dept Pharmacy and Pharmacology, University of Bath, UK
| | - Michael A Simpson
- Department of Medical and Molecular Genetics, School of Basic & Medical Biosciences, King's College London, London, UK
| | - Jonathan Packham
- Rheumatology Department, Haywood Hospital, Stoke on Trent, Midlands Partnership NHS Foundation Trust, UK.,Academic Unit of Population and Lifespan Sciences, University of Nottingham, University of Nottingham, UK
| | - Ross McManus
- Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Ireland
| | - Cem Gabay
- Division of Rheumatology, Department of Medicine, Geneva University Hospitals & Department of Pathology and Immunology, University of Geneva, Faculty of Medicine, Geneva, Switzerland
| | - Céline Lamacchia
- Division of Rheumatology, Geneva University Hospital, Geneva, Switzerland
| | - Michael J Nissen
- Division of Rheumatology, Geneva University Hospital, Geneva, Switzerland
| | - Matthew A Brown
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London, London, UK.,Genomics England, Charterhouse Square, London, UK
| | - Suzanne M M Verstappen
- NIHR Manchester Musculoskeletal Biomedical Research Unit, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, UK.,Centre for Epidemiology Versus Arthritis, Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Tjeerd Van Staa
- Health e-Research Centre, Health Data Research UK North, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, Manchester, UK
| | - Jonathan N Barker
- St John's Institute of Dermatology, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Catherine H Smith
- St John's Institute of Dermatology, Guys and St Thomas' Foundation Trust and Kings College London, London, UK
| | | | | | - Oliver FitzGerald
- UCD School of Medicine and Medical Sciences and Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Ireland
| | - Neil McHugh
- Royal National Hospital for Rheumatic Diseases and Dept Pharmacy and Pharmacology, University of Bath, UK
| | - Richard B Warren
- Dermatology Centre, Salford Royal NHS Foundation Trust, Manchester NIHR Biomedical Research Centre, University of Manchester, Manchester, UK
| | - John Bowes
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester, UK.,NIHR Manchester Musculoskeletal Biomedical Research Unit, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, UK
| | - Anne Barton
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester, UK.,NIHR Manchester Musculoskeletal Biomedical Research Unit, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, UK
| |
Collapse
|
34
|
Azhdari S, Saghi M, Alani B, Zare Rafie M, Kenarangi T, Nasrollahzadeh Sabet M, Pakzad B, Ghorashi T, Gholami M, Soosanabadi M. Assessment of the association between TNIP1 polymorphism with clinical features and risk of systemic lupus erythematosus. Lupus 2022; 31:903-909. [PMID: 35475371 DOI: 10.1177/09612033221094706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Over the past decades, TNIP1 has been identified as a strong risk locus in multiple genome-wide association studies (GWAS), spanning multiple populations and various autoimmune diseases. TNIP1 is a polyubiquitin-binding protein that works as a physiological inhibitor of NF-κB and maintains immune homeostasis. Some studies have confirmed that TNIP1 is downregulated in autoimmune diseases such as rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE). In the current study, for the first time, we evaluated the possible association between rs6889239 polymorphism in the TNIP1 gene with the risk and clinical characteristics of RA and SLE in the Iranian population. METHOD In this case-control study, 115 patients with RA, 115 patients with SLE, and 115 unrelated healthy subjects were enrolled to estimate rs6889239 genotypes with real-time PCR high resolution melting (HRM) method. RESULTS Our results demonstrated considerable associations between CC genotype and C allele of rs6889239 with augmented risk of SLE (OR for CC genotype= 2.23; 95%CI [1.175-4.307], OR for C allele= 1.84; 95%CI [1.254-2.720]). However, there was an insignificant association between genotypes and allele frequencies of rs6889239 with the occurrence risk of RA in the population under study (p > 0.05). Additionally, stratification analysis specified that the C allele in rs6889239 was linked with the incidence of renal involvement in SLE patients and lower age of onset in the RA group (p < 0.05). CONCLUSION These findings propose a significant association between TNIP1 polymorphism and higher risk of SLE and some clinical characteristics of RA and SLE.
Collapse
Affiliation(s)
- Sara Azhdari
- Department of Anatomy and Embryology, School of Medicine, 394237Bam University of Medical Sciences, Bam, Iran
| | - Mostafa Saghi
- School of Medicine, 162996AJA University of Medical Science, Tehran, Iran.,Fetal Health Research Center, Hope Generation Foundation, Tehran, Iran
| | - Behrang Alani
- Department of Applied Cell Sciences, Faculty of Medicine, 48462Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Zare Rafie
- Fetal Health Research Center, Hope Generation Foundation, Tehran, Iran.,School of Medicine, 48539Zanjan University of Medical Sciences, Zanjan, Iran
| | - Taiebe Kenarangi
- Student Research Committee, Faculty of Statistics, 48533University of Social Welfare and Rehabilitation Science, Tehran, Iran
| | | | - Bahram Pakzad
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, 108867Isfahan University of Medical Science, Isfahan, Iran
| | - Tahereh Ghorashi
- Department of Medical Genetics, 154203Semnan University of Medical Sciences, Semnan, Iran
| | - Milad Gholami
- Department of Biochemistry and Genetics, School of Medicine, 48412Arak University of Medical Sciences, Arak, Iran
| | - Mohsen Soosanabadi
- Department of Medical Genetics, 154203Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
35
|
Massy E, Pedini P, Pollet E, Martin M, Roudier J, Picard C, Balandraud N. Association study between HLA-A, -B, -C, -DRB1 alleles and Psoriatic arthritis in southern France. Hum Immunol 2022; 83:515-520. [DOI: 10.1016/j.humimm.2022.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 03/24/2022] [Accepted: 04/05/2022] [Indexed: 11/27/2022]
|
36
|
Laborde CM, Larzabal L, González-Cantero Á, Castro-Santos P, Díaz-Peña R. Advances of Genomic Medicine in Psoriatic Arthritis. J Pers Med 2022; 12:jpm12010035. [PMID: 35055350 PMCID: PMC8780979 DOI: 10.3390/jpm12010035] [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] [Received: 11/29/2021] [Revised: 12/16/2021] [Accepted: 12/28/2021] [Indexed: 12/13/2022] Open
Abstract
Psoriatic arthritis (PsA) is a common type of inflammatory arthritis found in up to 40% of patients with psoriasis. Although early diagnosis is important for reducing the risk of irreversible structural damage, there are no adequate screening tools for this purpose, and there are no clear markers of predisposition to the disease. Much evidence indicates that PsA disorder is complex and heterogeneous, where genetic and environmental factors converge to trigger inflammatory events and the development of the disease. Nevertheless, the etiologic events that underlie PsA are complex and not completely understood. In this review, we describe the existing data in PsA in order to highlight the need for further research in this disease to progress in the knowledge of its pathobiology and to obtain early diagnosis tools for these patients.
Collapse
Affiliation(s)
| | | | - Álvaro González-Cantero
- Department of Dermatology, Hospital Universitario Ramon y Cajal, 28034 Madrid, Spain;
- Faculty of Medicine, Universidad Francisco de Vitoria, Ctra. Pozuelo-Majadahonda, 28223 Pozuelo de Alarcón, 28034 Madrid, Spain
| | - Patricia Castro-Santos
- Immunology, Centro de Investigaciones Biomédicas (CINBIO), Universidad de Vigo, 36310 Vigo, Spain;
| | - Roberto Díaz-Peña
- Faculty of Health Sciences, Universidad Autónoma de Chile, Talca 3460000, Chile
- Correspondence: or ; Tel.: +34-981-955-073
| |
Collapse
|
37
|
Manou-Stathopoulou S, Lewis MJ. Diversity of NF-κB signalling and inflammatory heterogeneity in Rheumatic Autoimmune Disease. Semin Immunol 2021; 58:101649. [PMID: 36064646 DOI: 10.1016/j.smim.2022.101649] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Systemic Autoimmune Rheumatic Diseases, including Rheumatoid Arthritis, Systemic Lupus Erythematosus and Sjogren's syndrome, are characterised by a loss of immune tolerance and chronic inflammation. There is marked heterogeneity in clinical and molecular phenotypes in each condition, and the aetiology of these is unclear. NF-κB is an inducible transcription factor that is critical in the physiological inflammatory response, and which has been implicated in chronic inflammation. Genome-wide association studies have linked risk alleles related to the NF-κB pathway to the pathogenesis of multiple Systemic Autoimmune Rheumatic Diseases. This review describes how cell- and pathway-specific NF-κB activation contribute to the spectrum of clinical phenotypes and molecular pathotypes in rheumatic disease. Potential clinical applications are explored, including therapeutic interventions and utilisation of NF-κB as a biomarker of disease subtypes and treatment response.
Collapse
Affiliation(s)
- Sotiria Manou-Stathopoulou
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London, School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Myles J Lewis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London, School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
38
|
Advanced genomics and clinical phenotypes in psoriatic arthritis. Semin Immunol 2021; 58:101665. [PMID: 36307312 DOI: 10.1016/j.smim.2022.101665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Psoriatic Arthritis (PsA) is a complex polygenic inflammatory disease showing a variable musculoskeletal involvement in patients with skin psoriasis. PsA coexist in 25-40 % of patients with the dermatological manifestations, but PsA may also predate the appearance of psoriasis. Nonetheless, the immunopathogenesis of psoriasis and PsA manifest significant similarities, with a major role of the individual susceptibility in both cases. Genome wide association studies (GWAS) identified several genes/loci associated with the risk to develop PsA, both dependent and independent of psoriasis. The major challenge is thus represented by the need to translate the identification of functional polymorphisms and other genetics findings into biological mechanisms along with the identification of novel putative drug targets. A functional genomics approach aims to increase GWAS power and recent evidence supports the use of a multilayer process, including eQTL, methylome, chromatin conformation analysis and genome editing to discover novel genes that can be affected by disease-associated variants, such as PsA. The available data have considered PsA as a unique homogeneous clinical entity while the clinical experience supports a wide variability of skin and joint manifestations coexisting in diverse patients with different mechanisms underlying the musculoskeletal and dermatological domains. A better discrimination of the patient features is encouraged by the limited data on functional genomics. We provide herein a review of the latest findings on PsA functional genomics highlighting the exciting developments in the field and how these might lead to a better understanding of gene regulation underpinning disease mechanisms and ultimately refine clinical phenotyping.
Collapse
|
39
|
Kingsmore KM, Puglisi CE, Grammer AC, Lipsky PE. An introduction to machine learning and analysis of its use in rheumatic diseases. Nat Rev Rheumatol 2021; 17:710-730. [PMID: 34728818 DOI: 10.1038/s41584-021-00708-w] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2021] [Indexed: 02/07/2023]
Abstract
Machine learning (ML) is a computerized analytical technique that is being increasingly employed in biomedicine. ML often provides an advantage over explicitly programmed strategies in the analysis of multidimensional information by recognizing relationships in the data that were not previously appreciated. As such, the use of ML in rheumatology is increasing, and numerous studies have employed ML to classify patients with rheumatic autoimmune inflammatory diseases (RAIDs) from medical records and imaging, biometric or gene expression data. However, these studies are limited by sample size, the accuracy of sample labelling, and absence of datasets for external validation. In addition, there is potential for ML models to overfit or underfit the data and, thereby, these models might produce results that cannot be replicated in an unrelated dataset. In this Review, we introduce the basic principles of ML and discuss its current strengths and weaknesses in the classification of patients with RAIDs. Moreover, we highlight the successful analysis of the same type of input data (for example, medical records) with different algorithms, illustrating the potential plasticity of this analytical approach. Altogether, a better understanding of ML and the future application of advanced analytical techniques based on this approach, coupled with the increasing availability of biomedical data, may facilitate the development of meaningful precision medicine for patients with RAIDs.
Collapse
Affiliation(s)
| | | | - Amrie C Grammer
- AMPEL BioSolutions and RILITE Research Institute, Charlottesville, VA, USA
| | - Peter E Lipsky
- AMPEL BioSolutions and RILITE Research Institute, Charlottesville, VA, USA
| |
Collapse
|
40
|
Kubanov AA, Karamova AE, Chikin VV, Verbenko DA, Znamenskaya LF, Artamonova OG. Genetic markers for psoriatic arthritis in patients with psoriasis. Part I: non-HLA genes. VESTNIK DERMATOLOGII I VENEROLOGII 2021. [DOI: 10.25208/vdv1260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Psoriatic arthritis often develops in patients with psoriasis and can lead to joint deformity, stiffness, dysfunction, and disability. Psoriatic arthritis is a polygenic disease. and the issue of personalizing the prognosis of its development can only be resolved taking into account the variability of plenty genomic loci associated with the development of the disease. The personification of the prognosis of the disease can be solved taking into account the variability of the set of genomic loci with which its development is associated. The review examines genomic polymorphisms associated with the development of psoriatic arthritis not psoriasis, except of HLA polymorphisms. Genome regions containing polymorphisms, allelic variants of which are associated both with the development of psoriatic arthritis and reducing the likelihood of its occurrence, are described. It has been reported that the predisposition to the development of psoriatic arthritis in patients with psoriasis is determined by genes encoding proteins involved in inflammation and bone metabolism.
Collapse
|
41
|
Chia AYT, Ang GWX, Chan ASY, Chan W, Chong TKY, Leung YY. Managing Psoriatic Arthritis With Inflammatory Bowel Disease and/or Uveitis. Front Med (Lausanne) 2021; 8:737256. [PMID: 34604268 PMCID: PMC8481670 DOI: 10.3389/fmed.2021.737256] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 08/20/2021] [Indexed: 12/15/2022] Open
Abstract
Psoriatic arthritis (PsA) is a chronic inflammatory disease that presents with psoriasis (PsO), peripheral and axial arthropathy. The heterogeneity of disease presentation leads to the term "psoriatic disease (PsD)" which is thought to better encompass the range of clinical manifestations. PsA is associated with several comorbidities such as cardiovascular diseases, metabolic syndrome and other extra-articular manifestations including uveitis, and inflammatory bowel disease (IBD). While novel therapeutics are being developed following advances in our understanding of the pathogenesis of the disease, the diverse combinations of PsA with its various comorbidities still pose a clinical challenge in managing patients with PsA. This article reviews our current understanding of the pathogenesis of PsA and how various pathways in the pathogenesis lead to the two comorbid extra-articular manifestations - uveitis and IBD. We also review current evidence of treatment strategies in managing patients with PsA with comorbidities of uveitis and/or IBD.
Collapse
Affiliation(s)
- Alfred Yu Ting Chia
- Duke-NUS Medical School, Singapore, Singapore
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Duke-NUS Medical School, Singapore, Singapore
| | - Gladys Wei Xin Ang
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Duke-NUS Medical School, Singapore, Singapore
| | - Anita Sook Yee Chan
- Duke-NUS Medical School, Singapore, Singapore
- Singapore National Eye Center and Singapore Eye Research Center, Singapore, Singapore
| | - Webber Chan
- Department of Gastroenterology and Hepatology, Singapore General Hospital, Singapore, Singapore
| | | | - Ying Ying Leung
- Duke-NUS Medical School, Singapore, Singapore
- Department of Rheumatology and Immunology, Singapore General Hospital, Singapore, Singapore
| |
Collapse
|
42
|
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
|
43
|
Yan D, Gudjonsson JE, Le S, Maverakis E, Plazyo O, Ritchlin C, Scher JU, Singh R, Ward NL, Bell S, Liao W. New Frontiers in Psoriatic Disease Research, Part I: Genetics, Environmental Triggers, Immunology, Pathophysiology, and Precision Medicine. J Invest Dermatol 2021; 141:2112-2122.e3. [PMID: 34303522 PMCID: PMC8384663 DOI: 10.1016/j.jid.2021.02.764] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 02/07/2023]
Abstract
Psoriasis is a chronic inflammatory condition characterized by systemic immune dysregulation. Over the past several years, advances in genetics, microbiology, immunology, and mouse models have revealed the complex interplay between the heritable and microenvironmental factors that drive the development of psoriatic inflammation. In the first of this two-part review series, the authors will discuss the newest insights into the pathogenesis of psoriatic disease and highlight how the evolution of these scientific fields has paved the way for a more personalized approach to psoriatic disease treatment.
Collapse
Affiliation(s)
- Di Yan
- Ronald O. Perelman Department of Dermatology, NYU Langone Health, New York, New York, USA
| | | | - Stephanie Le
- Department of Dermatology, University of California, Davis, Sacramento, California, USA
| | - Emanual Maverakis
- Department of Dermatology, University of California, Davis, Sacramento, California, USA
| | - Olesya Plazyo
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | - Christopher Ritchlin
- Center for Musculoskeletal Research, Division of Allergy, Immunology and Rheumatology, University of Rochester School of Medicine & Dentistry, Rochester, New York, USA
| | - Jose U Scher
- Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, New York, USA
| | - Roopesh Singh
- Department of Nutrition, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Nicole L Ward
- Department of Nutrition, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA; Department of Dermatology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Stacie Bell
- National Psoriasis Foundation, Portland, Oregon, USA
| | - Wilson Liao
- UCSF Department of Dermatology, University of California San Francisco, San Francisco, California, USA.
| |
Collapse
|
44
|
Hromadová D, Elewaut D, Inman RD, Strobl B, Gracey E. From Science to Success? Targeting Tyrosine Kinase 2 in Spondyloarthritis and Related Chronic Inflammatory Diseases. Front Genet 2021; 12:685280. [PMID: 34290741 PMCID: PMC8287328 DOI: 10.3389/fgene.2021.685280] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/02/2021] [Indexed: 12/16/2022] Open
Abstract
Spondyloarthritis (SpA) is a family of inflammatory arthritic diseases, which includes the prototypes of psoriatic arthritis and ankylosing spondylitis. SpA is commonly associated with systemic inflammatory diseases, such as psoriasis and inflammatory bowel disease. Immunological studies, murine models and the genetics of SpA all indicate a pathogenic role for the IL-23/IL-17 axis. Therapeutics targeting the IL-23/IL-17 pathway are successful at providing symptomatic relief, but may not provide complete protection against progression of arthritis. Thus there is still tremendous interest in the discovery of novel therapeutic targets for SpA. Tyrosine kinase 2 (TYK2) is a member of the Janus kinases, which mediate intracellular signaling of cytokines via signal transducer and activator of transcription (STAT) activation. TYK2 plays a crucial role in mediating IL-23 receptor signaling and STAT3 activation. A plethora of natural mutations in and around TYK2 have provided a wealth of data to associate this kinase with autoimmune/autoinflammatory diseases in humans. Induced and natural mutations in murine Tyk2 largely support human data; however, key inter-species differences exist, which means extrapolation of data from murine models to humans needs to be done with caution. Despite these reservations, novel selective TYK2 inhibitors are now proving successful in advanced clinical trials of inflammatory diseases. In this review, we will discuss TYK2 from basic biology to therapeutic targeting, with an emphasis on studies in SpA. Seminal studies uncovering the basic science of TYK2 have provided sound foundations for targeting it in SpA and related inflammatory diseases. TYK2 inhibitors may well be the next blockbuster therapeutic for SpA.
Collapse
Affiliation(s)
- Dominika Hromadová
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Dirk Elewaut
- Molecular Immunology and Inflammation Unit, VIB Centre for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Rheumatology, Ghent University Hospital, Ghent, Belgium
| | - Robert D. Inman
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada
- Departments of Medicine and Immunology, University of Toronto, Toronto, ON, Canada
| | - Birgit Strobl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Eric Gracey
- Molecular Immunology and Inflammation Unit, VIB Centre for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Rheumatology, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
45
|
Leijten EF, van Kempen TS, Olde Nordkamp MA, Pouw JN, Kleinrensink NJ, Vincken NL, Mertens J, Balak DMW, Verhagen FH, Hartgring SA, Lubberts E, Tekstra J, Pandit A, Radstake TR, Boes M. Tissue-Resident Memory CD8+ T Cells From Skin Differentiate Psoriatic Arthritis From Psoriasis. Arthritis Rheumatol 2021; 73:1220-1232. [PMID: 33452865 PMCID: PMC8362143 DOI: 10.1002/art.41652] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 01/07/2021] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To compare immune cell phenotype and function in psoriatic arthritis (PsA) versus psoriasis in order to better understand the pathogenesis of PsA. METHODS In-depth immunophenotyping of different T cell and dendritic cell subsets was performed in patients with PsA, psoriasis, or axial spondyloarthritis and healthy controls. Subsequently, we analyzed cells from peripheral blood, synovial fluid (SF), and skin biopsy specimens using flow cytometry, along with high-throughput transcriptome analyses and functional assays on the specific cell populations that appeared to differentiate PsA from psoriasis. RESULTS Compared to healthy controls, the peripheral blood of patients with PsA was characterized by an increase in regulatory CD4+ T cells and interleukin-17A (IL-17A) and IL-22 coproducing CD8+ T cells. One population specifically differentiated PsA from psoriasis: i.e., CD8+CCR10+ T cells were enriched in PsA. CD8+CCR10+ T cells expressed high levels of DNAX accessory molecule 1 and were effector memory cells that coexpressed skin-homing receptors CCR4 and cutaneous lymphocyte antigen. CD8+CCR10+ T cells were detected under inflammatory and homeostatic conditions in skin, but were not enriched in SF. Gene profiling further revealed that CD8+CCR10+ T cells expressed GATA3, FOXP3, and core transcriptional signature of tissue-resident memory T cells, including CD103. Specific genes, including RORC, IFNAR1, and ERAP1, were up-regulated in PsA compared to psoriasis. CD8+CCR10+ T cells were endowed with a Tc2/22-like cytokine profile, lacked cytotoxic potential, and displayed overall regulatory function. CONCLUSION Tissue-resident memory CD8+ T cells derived from the skin are enhanced in the circulation of patients with PsA compared to patients with psoriasis alone. This may indicate that aberrances in cutaneous tissue homeostasis contribute to arthritis development.
Collapse
MESH Headings
- Adult
- Aminopeptidases/genetics
- Antigens, CD/genetics
- Antigens, Differentiation, T-Lymphocyte/immunology
- Arthritis, Psoriatic/genetics
- Arthritis, Psoriatic/immunology
- Arthritis, Psoriatic/pathology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Case-Control Studies
- Female
- Forkhead Transcription Factors/genetics
- GATA3 Transcription Factor/genetics
- Gene Expression Profiling
- High-Throughput Nucleotide Sequencing
- Humans
- Immunologic Memory/immunology
- Immunophenotyping
- Integrin alpha Chains/genetics
- Interleukin-17/immunology
- Interleukins/immunology
- Male
- Middle Aged
- Minor Histocompatibility Antigens/genetics
- Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
- Oligosaccharides/metabolism
- Psoriasis/genetics
- Psoriasis/immunology
- Psoriasis/pathology
- Receptor, Interferon alpha-beta/genetics
- Receptors, CCR10/metabolism
- Receptors, CCR4/metabolism
- Sialyl Lewis X Antigen/analogs & derivatives
- Sialyl Lewis X Antigen/metabolism
- Skin/immunology
- Skin/pathology
- Spondylarthropathies/genetics
- Spondylarthropathies/immunology
- Spondylarthropathies/pathology
- Synovial Fluid/cytology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Interleukin-22
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jorre Mertens
- University Medical Center UtrechtUtrechtThe Netherlands
| | | | | | | | - Erik Lubberts
- Erasmus University Medical CenterRotterdamThe Netherlands
| | | | | | | | - Marianne Boes
- University Medical Center UtrechtUtrechtThe Netherlands
| |
Collapse
|
46
|
Tang J. Immunogenetic determinants of heterosexual HIV-1 transmission: key findings and lessons from two distinct African cohorts. Genes Immun 2021; 22:65-74. [PMID: 33934119 PMCID: PMC8225584 DOI: 10.1038/s41435-021-00130-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 04/05/2021] [Accepted: 04/15/2021] [Indexed: 02/03/2023]
Abstract
Immunogenetic studies in the past three decades have uncovered a broad range of human genetic factors that seem to influence heterosexual HIV-1 transmission in one way or another. In our own work that jointly evaluated both genetic and nongenetic factors in two African cohorts of cohabiting, HIV-1-discordant couples (donor and recipient pairs) at risk of transmission during quarterly follow-up intervals, relatively consistent findings have been seen with three loci (IL19, HLA-A, and HLA-B), although the effect size (i.e., odds ratio or hazards ratio) of each specific variant was quite modest. These studies offered two critical lessons that should benefit future research on sexually transmitted infections. First, in donor partners, immunogenetic factors (e.g., HLA-B*57 and HLA-A*36:01) that operate directly through HIV-1 viral load or indirectly through genital coinfections are equally important. Second, thousands of single-nucleotide polymorphisms previously recognized as "causal" factors for human autoimmune disorders did not appear to make much difference, which is somewhat puzzling as these variants are predicted or known to influence the expression of many immune response genes. Replicating these observations in additional cohorts is no longer feasible as the field has shifted its focus to early diagnosis, universal treatment, and active management of comorbidities.
Collapse
Affiliation(s)
- Jianming Tang
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
47
|
|
48
|
O'Rielly DD, Rahman P. Clinical and molecular significance of genetic loci associated with psoriatic arthritis. Best Pract Res Clin Rheumatol 2021; 35:101691. [PMID: 34020887 DOI: 10.1016/j.berh.2021.101691] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Psoriatic arthritis (PsA) is caused by a combination of environmental and multiple genetic factors, with clear evidence for a strong genetic basis. The remarkable accumulation of knowledge gained from genetic, pharmacogenetic, and therapeutic response of biologic agents in PsA has fundamentally changed and advanced our understanding of disease pathogenesis and has identified key signalling pathways. However, only one-quarter of the genetic contribution of PsA has been accounted for; and dissecting the genetic contributors of the cutaneous disease from those that would identify joint disease has been challenging. More importantly, the clinical utility of multiple proposed loci is unclear. In this review, we summarize the potential clinical relevance from established genetic associations and provide insight on the proposed molecular pathways that arise from these associations.
Collapse
Affiliation(s)
- Darren D O'Rielly
- Faculty of Medicine, Memorial University, Craig L Dobbin Genetics Research Centre, Suite 3M500, 300 Prince Philip Drive, St. John's, NL, A1B3V6, Canada
| | - Proton Rahman
- St. Clare's Mercy Hospital, 154 LeMarchant Rd, St. John's, Newfoundland, A1C5B8, Canada.
| |
Collapse
|
49
|
Lugar CW, Clarke CA, Morphy R, Rudyk H, Sapmaz S, Stites RE, Vaught GM, Furness K, Broughton HB, Durst GL, Clawson DK, Stout SL, Guo SY, Durbin JD, Stayrook KR, Edmondson DD, Kikly K, New NE, Bina HA, Chambers MG, Shetler P, Chang WY, Chang VCY, Barr R, Gough WH, Steele JP, Getman B, Patel N, Mathes BM, Richardson TI. Defining Target Engagement Required for Efficacy In Vivo at the Retinoic Acid Receptor-Related Orphan Receptor C2 (RORγt). J Med Chem 2021; 64:5470-5484. [PMID: 33852312 DOI: 10.1021/acs.jmedchem.0c01918] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The Th17 pathway has been implicated in autoimmune diseases. The retinoic acid receptor-related orphan receptor C2 (RORγt) is a master regulator of Th17 cells and controls the expression of IL-17A. RORγt is expressed primarily in IL-17A-producing lymphoid cells. Here we describe a virtual screen of the ligand-binding pocket and subsequent screen in a binding assay that identified the 1-benzyl-4',5'-dihydrospiro[piperidine-4,7'-thieno[2,3-c]pyran]-2'-carboxamide scaffold as a starting point for optimization of binding affinity and functional activity guided by structure-based design. Compound 12 demonstrated activity in a mouse PK/PD model and efficacy in an inflammatory arthritis mouse model that were used to define the level and duration of target engagement required for efficacy in vivo. Further optimization to improve ADME and physicochemical properties with guidance from simulations and modeling provided compound 22, which is projected to achieve the level and duration of target engagement required for efficacy in the clinic.
Collapse
Affiliation(s)
- Charles W Lugar
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Christian A Clarke
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Richard Morphy
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Helene Rudyk
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Selma Sapmaz
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Ryan E Stites
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Grant M Vaught
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Kelly Furness
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Howard B Broughton
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Greg L Durst
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - David K Clawson
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Stephanie L Stout
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Sherry Y Guo
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Jim D Durbin
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Keith R Stayrook
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Denise D Edmondson
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Kristy Kikly
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Nicole E New
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Holly A Bina
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Mark G Chambers
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Pamela Shetler
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - William Y Chang
- Lilly Biotechnology Center, Eli Lilly and Company, San Diego, California 92121, United States
| | - Veavi Ching-Yun Chang
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Rob Barr
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Wendy H Gough
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Jimmy P Steele
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Brian Getman
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Nita Patel
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Brian M Mathes
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Timothy I Richardson
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| |
Collapse
|
50
|
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
|