1
|
Chaitidis N, Papadopoulou Z, Varvara ST, Panagiotidis M, Katsigianni I, Sakellariou GT. Paradoxical psoriasis induced by IL-17 inhibitors: a case series of patients with axial spondyloarthritis and a systematic literature review. Rheumatol Int 2024; 44:2659-2668. [PMID: 38914777 DOI: 10.1007/s00296-024-05647-9] [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: 02/17/2024] [Accepted: 06/08/2024] [Indexed: 06/26/2024]
Abstract
Following the market authorization of interleukin (IL)-17 inhibitors, a growing number of cases of IL-17 inhibitor-induced paradoxical psoriasis (PsO) have been reported. Our objectives were to present two cases of IL-17 inhibitor-induced paradoxical PsO and to systematically review the literature for similar cases, summarizing and presenting the relevant data. A systematic literature review of previously presented cases of paradoxical PsO induced by IL-17 inhibitors was conducted. We presented two patients with axial spondyloarthritis (axSpA) and paradoxical PsO induced by secukinumab (SEC). One patient's psoriatic lesions responded well to adjuvant topical treatment, while the other patient required a combination of topical treatment and cyclosporine Α for successful treatment. SEC was continued in both cases. We also identified 35 patients with IL-17 inhibitor-induced paradoxical PsO in the literature review. The most frequent types of paradoxical PsO were palmoplantar pustular and plaque PsO, while the median latency period was 11 weeks. Approximately one-third of patients continued IL-17 inhibitor treatment with adjunctive therapy, primarily topical, which produced satisfactory results in most patients. Almost two-thirds of the patients discontinued the IL-17 inhibitor, with the majority of patients switching to another biological agent with a different mechanism of action or initiating other systemic antipsoriatic treatments, resulting in mainly satisfactory outcomes. Therefore, paradoxical PsO induced by IL-17 inhibitors appears to respond well in both patients who continue IL-17 inhibitors with adjunctive treatment and those who discontinue IL-17 inhibitors while switching to a different class of biological agent or initiating other systemic antipsoriatic treatments.
Collapse
Affiliation(s)
- Nikolaos Chaitidis
- Department of Dermatology and Venereology, 424 General Military Training Hospital, Thessaloniki, Hellenic Republic, Greece.
| | - Zoi Papadopoulou
- 3rd Department of Pediatrics, Hippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Hellenic Republic, Greece
| | - Stavritsa Taxiarchoula Varvara
- Department of Dermatology and Venereology, 424 General Military Training Hospital, Thessaloniki, Hellenic Republic, Greece
- 2nd Department of Internal Medicine, 424 General Military Training Hospital, Thessaloniki, Hellenic Republic, Greece
| | - Michail Panagiotidis
- 3rd Department of Internal Medicine, Papageorgiou General Hospital, Thessaloniki, Hellenic Republic, Greece
| | - Ioanna Katsigianni
- Department of Rheumatology, 424 General Military Training Hospital, Thessaloniki, Hellenic Republic, Greece
| | - Grigorios T Sakellariou
- Department of Rheumatology, 424 General Military Training Hospital, Thessaloniki, Hellenic Republic, Greece
| |
Collapse
|
2
|
Su QY, Gao HY, Duan YR, Luo J, Wang WZ, Qiao XC, Zhang SX. The immunologic role of IL-23 in psoriatic arthritis: a potential therapeutic target. Expert Opin Biol Ther 2024; 24:1119-1132. [PMID: 39230202 DOI: 10.1080/14712598.2024.2401148] [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: 03/10/2024] [Revised: 07/25/2024] [Accepted: 09/02/2024] [Indexed: 09/05/2024]
Abstract
INTRODUCTION Psoriatic arthritis (PsA) is a debilitating chronic condition characterized by inflammation of the joints, bones, enthesis, and skin. The pivotal role of interleukin-23 (IL-23) in the pathogenesis of PsA has become increasingly evident. This proinflammatory cytokine is markedly elevated in patients with PsA, suggesting its potential as a therapeutic target. Consequently, IL-23 inhibitors have emerged as promising first-line biologic treatments for PsA. AREAS COVERED This review delves into the immunopathogenic mechanisms of IL-23 at the cellular and molecular levels in PsA. Furthermore, it provides the recent efficacy and safety profiles of IL-23 inhibitors. We conducted a literature search in PubMed for the following terms: 'IL-23 and psoriatic arthritis,' 'Ustekinumab,' 'Guselkumab,' 'Risankizumab,' and 'Tildrakizumab.' In addition, we retrieved clinical trials involving IL-23 inhibitors registered in ClinicalTrials.gov, EudraCT, and ICTRP. EXPERT OPINION Despite the promising outcomes observed with IL-23 inhibitors, several challenges persist. The long-term effects of these agents require further investigation through prospective studies, and their limited accessibility worldwide necessitates urgent attention. Additionally, ongoing research is warranted to explore other potential drug targets within the IL-23/IL-23 R axis. The development of reliable biomarkers could greatly enhance early detection, tailored management strategies, and personalized treatment approaches for patients with PsA.
Collapse
Affiliation(s)
- Qin-Yi Su
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Heng-Yan Gao
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Yue-Ru Duan
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Jing Luo
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Wei-Ze Wang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Xi-Chao Qiao
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Sheng-Xiao Zhang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi medical university, Taiyuan, China
| |
Collapse
|
3
|
Tang Y, Zhuo D, Yu Y, Pu W, Ma Y, Zhang Y, Huang Y, Zhang Q, Tang K, Meng C, Yang D, Bai L, He D, Jin L, Zou H, Xu H, Zhu Q, Wang J, Chen Y, Liu J. Single-cell RNA sequencing reveals the CRTAC1 + population actively contributes to the pathogenesis of spinal ligament degeneration by SPP1 + macrophage. Aging Cell 2024:e14320. [PMID: 39158018 DOI: 10.1111/acel.14320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/12/2024] [Accepted: 08/07/2024] [Indexed: 08/20/2024] Open
Abstract
Degenerative spinal stenosis is a chronic disease that affects the spinal ligaments and associated bones, resulting in back pain and disorders of the limbs among the elderly population. There are few preventive strategies for such ligament degeneration. We here aimed to establish a comprehensive transcriptomic atlas of ligament tissues to identify high-priority targets for pharmaceutical treatment of ligament degeneration. Here, single-cell RNA sequencing was performed on six degenerative ligaments and three traumatic ligaments to understand tissue heterogeneity. After stringent quality control, high-quality data were obtained from 32,014 cells. Distinct cell clusters comprising stromal and immune cells were identified in ligament tissues. Among them, we noted that collagen degradation associated with CTHRC1+ fibroblast-like cells and calcification linked to CRTAC1+ chondrocyte-like cells were key features of ligament degeneration. SCENIC analysis and further experiments identified ATF3 as a key transcription factor regulating the pathogenesis of CRTAC1+ chondrocyte-like cells. Typically, immune cells infiltrate localized organs, causing tissue damage. In our study, myeloid cells were found to be inflammatory-activated, and SPP1+ macrophages were notably enriched in degenerative ligaments. Further exploration via CellChat analysis demonstrated a robust interaction between SPP1+ macrophages and CRTAC1+ chondrocyte-like cells. Activated by SPP1, ATF3 propels the CRTAC1/MGP/CLU axis, fostering ligament calcification. Our unique resource provides novel insights into possible mechanisms underlying ligament degeneration, the target cell types, and molecules that are expected to mitigate degenerative spinal ligament. We also highlight the role of immune regulation in ligament degeneration and calcification, enhancing our understanding of this disease.
Collapse
Affiliation(s)
- Yulong Tang
- Shanghai Key Laboratory of Vascular Lesions and Remodeling, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, and Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, China
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China
| | - Dachun Zhuo
- Shanghai Key Laboratory of Vascular Lesions and Remodeling, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, and Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, China
| | - Yuexin Yu
- Shanghai Key Laboratory of Vascular Lesions and Remodeling, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, and Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, China
| | - Weilin Pu
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China
- Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Guangzhou, China
| | - Yanyun Ma
- Ministry of Education Key Laboratory of Contemporary Anthropology, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai, China
- Research Unit of Dissecting the Population Genetics and Developing New Technologies for Treatment and Prevention of Skin Phenotypes and Dermatological Diseases (2019RU058), Chinese Academy of Medical Sciences, Beijing, China
| | - Yuting Zhang
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China
| | - Yan Huang
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China
| | - Qing Zhang
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China
| | - Kunhai Tang
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China
| | - Chen Meng
- Shanghai Key Laboratory of Vascular Lesions and Remodeling, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, and Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, China
| | - Di Yang
- Shanghai Key Laboratory of Vascular Lesions and Remodeling, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, and Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, China
| | - Lu Bai
- Shanghai Key Laboratory of Vascular Lesions and Remodeling, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, and Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, China
| | - Dongyi He
- Institute of Arthritis Research, Shanghai Academy of Chinese Medical Sciences, Guanghua Integrative Medicine Hospital, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China
- Research Unit of Dissecting the Population Genetics and Developing New Technologies for Treatment and Prevention of Skin Phenotypes and Dermatological Diseases (2019RU058), Chinese Academy of Medical Sciences, Beijing, China
| | - Hejian Zou
- Division of Rheumatology, Huashan Hospital, and Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Huji Xu
- Department of Rheumatology and Immunology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Qi Zhu
- Institute of Arthritis Research, Shanghai Academy of Chinese Medical Sciences, Guanghua Integrative Medicine Hospital, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China
- Research Unit of Dissecting the Population Genetics and Developing New Technologies for Treatment and Prevention of Skin Phenotypes and Dermatological Diseases (2019RU058), Chinese Academy of Medical Sciences, Beijing, China
- Division of Rheumatology, Huashan Hospital, and Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Yuanyuan Chen
- Orthopedic Department, Shanghai Sixth People's Hospital, Shanghai Jiaotong University Affiliated, Shanghai, China
- Orthopaedic Department, People's Hospital of Shigatse City, Shigatse, China
| | - Jing Liu
- Shanghai Key Laboratory of Vascular Lesions and Remodeling, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, and Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, China
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China
| |
Collapse
|
4
|
Zhao M, Cheng Y, Gao J, Zhou F. Single-cell mass cytometry in immunological skin diseases. Front Immunol 2024; 15:1401102. [PMID: 39081313 PMCID: PMC11286489 DOI: 10.3389/fimmu.2024.1401102] [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: 03/14/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Immune-related skin diseases represent a collective of dermatological disorders intricately linked to dysfunctional immune system processes. These conditions are primarily characterized by an immoderate activation of the immune system or deviant immune responses, involving diverse immune components including immune cells, antibodies, and inflammatory mediators. However, the precise molecular dysregulation underlying numerous individual cases of these diseases and unique subsets respond under disease conditions remains elusive. Comprehending the mechanisms and determinants governing the homeostasis and functionality of diseases could offer potential therapeutic opportunities for intervention. Mass cytometry enables precise and high-throughput quantitative measurement of proteins within individual cells by utilizing antibodies labeled with rare heavy metal isotopes. Imaging mass cytometry employs mass spectrometry to obtain spatial information on cell-to-cell interactions within tissue sections, simultaneously utilizing more than 40 markers. The application of single-cell mass cytometry presents a unique opportunity to conduct highly multiplexed analysis at the single-cell level, thereby revolutionizing our understanding of cell population heterogeneity and hierarchy, cellular states, multiplexed signaling pathways, proteolysis products, and mRNA transcripts specifically in the context of many autoimmune diseases. This information holds the potential to offer novel approaches for the diagnosis, prognostic assessment, and monitoring responses to treatment, thereby enriching our strategies in managing the respective conditions. This review summarizes the present-day utilization of single-cell mass cytometry in studying immune-related skin diseases, highlighting its advantages and limitations. This technique will become increasingly prevalent in conducting extensive investigations into these disorders, ultimately yielding significant contributions to their accurate diagnosis and efficacious therapeutic interventions.
Collapse
Affiliation(s)
- Mingming Zhao
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Yuqi Cheng
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Jinping Gao
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Fusheng Zhou
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| |
Collapse
|
5
|
Yang KL, Mullins BJ, Lejeune A, Ivanova E, Shin J, Bajwa S, Possemato R, Cadwell K, Scher JU, Koralov SB. Mitigation of Osteoclast-Mediated Arthritic Bone Remodeling By Short Chain Fatty Acids. Arthritis Rheumatol 2024; 76:647-659. [PMID: 37994265 PMCID: PMC10965381 DOI: 10.1002/art.42765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 10/24/2023] [Accepted: 11/13/2023] [Indexed: 11/24/2023]
Abstract
OBJECTIVE The objective for this study was to evaluate the effects of short chain fatty acids (SCFAs) on arthritic bone remodeling. METHODS We treated a recently described preclinical murine model of psoriatic arthritis (PsA), R26STAT3Cstopfl/fl CD4Cre mice, with SCFA-supplemented water. We also performed in vitro osteoclast differentiation assays in the presence of serum-level SCFAs to evaluate the direct impact of these microbial metabolites on maturation and function of osteoclasts. We further characterized the molecular mechanism of SCFAs by transcriptional analysis. RESULTS The osteoporosis condition in R26STAT3Cstopfl/fl CD4Cre animals is attributed primarily to robust osteoclast differentiation driven by an expansion of osteoclast progenitor cells (OCPs), accompanied by impaired osteoblast development. We show that SCFA supplementation can rescue the osteoporosis phenotype in this model of PsA. Our in vitro experiments revealed an inhibitory effect of the SCFAs on osteoclast differentiation, even at very low serum concentrations. This suppression of osteoclast differentiation enabled SCFAs to impede osteoporosis development in R26STAT3Cstopfl/fl CD4Cre mice. Further interrogation revealed that bone marrow-derived OCPs from diseased mice expressed a higher level of SCFA receptors than those of control mice and that the progenitor cells in the bone marrow of SCFA-treated mice presented a modified transcriptomic landscape, suggesting a direct impact of SCFAs on bone marrow progenitors in the context of osteoporosis. CONCLUSION We demonstrated how gut microbiota-derived SCFAs can regulate distal pathology (ie, osteoporosis) and identified a potential therapeutic option for restoring bone density in rheumatic disease, further highlighting the critical role of the gut-bone axis in these disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ken Cadwell
- NYU Langone Health, New York, NY
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Jose U. Scher
- New York University School of Medicine, New York, NY
- Judith and Stewart Colton Center for Autoimmunity, NYU Langone Health, New York, NY
| | | |
Collapse
|
6
|
Baumgartner F, Bamopoulos SA, Faletti L, Hsiao HJ, Holz M, Gonzalez-Menendez I, Solé-Boldo L, Horne A, Gosavi S, Özerdem C, Singh N, Liebig S, Ramamoorthy S, Lehmann M, Demel U, Kühl AA, Wartewig T, Ruland J, Wunderlich FT, Schick M, Walther W, Rose-John S, Haas S, Quintanilla-Martinez L, Feske S, Ehl S, Glauben R, Keller U. Activation of gp130 signaling in T cells drives T H17-mediated multi-organ autoimmunity. Sci Signal 2024; 17:eadc9662. [PMID: 38377177 DOI: 10.1126/scisignal.adc9662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 01/31/2024] [Indexed: 02/22/2024]
Abstract
The IL-6-gp130-STAT3 signaling axis is a major regulator of inflammation. Activating mutations in the gene encoding gp130 and germline gain-of-function mutations in STAT3 (STAT3GOF) are associated with multi-organ autoimmunity, severe morbidity, and adverse prognosis. To dissect crucial cellular subsets and disease biology involved in activated gp130 signaling, the gp130-JAK-STAT3 axis was constitutively activated using a transgene, L-gp130, specifically targeted to T cells. Activating gp130 signaling in T cells in vivo resulted in fatal, early onset, multi-organ autoimmunity in mice that resembled human STAT3GOF disease. Female mice had more rapid disease progression than male mice. On a cellular level, gp130 signaling induced the activation and effector cell differentiation of T cells, promoted the expansion of T helper type 17 (TH17) cells, and impaired the activity of regulatory T cells. Transcriptomic profiling of CD4+ and CD8+ T cells from these mice revealed commonly dysregulated genes and a gene signature that, when applied to human transcriptomic data, improved the segregation of patients with transcriptionally diverse STAT3GOF mutations from healthy controls. The findings demonstrate that increased gp130-STAT3 signaling leads to TH17-driven autoimmunity that phenotypically resembles human STAT3GOF disease.
Collapse
Affiliation(s)
- Francis Baumgartner
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité (Junior) (Digital) Clinician Scientist Program, 10178 Berlin, Germany
- German Cancer Consortium (DKTK), partner site Berlin, a partnership between DKFZ and Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Stefanos A Bamopoulos
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité (Junior) (Digital) Clinician Scientist Program, 10178 Berlin, Germany
- German Cancer Consortium (DKTK), partner site Berlin, a partnership between DKFZ and Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Laura Faletti
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, University Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Hsiang-Jung Hsiao
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Maximilian Holz
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
- German Cancer Consortium (DKTK), partner site Berlin, a partnership between DKFZ and Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Irene Gonzalez-Menendez
- Institute of Pathology and Neuropathology, Comprehensive Cancer Center, Eberhard Karls University of Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," Eberhard Karls University, 72076 Tübingen, Germany
- German Cancer Consortium (DKTK), partner site Tübingen, a partnership between DKFZ and Eberhard Karls University of Tübingen, 72076 Tübingen, Germany
| | - Llorenç Solé-Boldo
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 10115 Berlin, Germany
| | - Arik Horne
- German Cancer Consortium (DKTK), partner site Berlin, a partnership between DKFZ and Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 10115 Berlin, Germany
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) Heidelberg, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Sanket Gosavi
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Ceren Özerdem
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 10115 Berlin, Germany
| | - Nikita Singh
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Sven Liebig
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Senthilkumar Ramamoorthy
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, University Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center - University of Freiburg, 79110 Freiburg, Germany
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Malte Lehmann
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- iPATH.Berlin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Benjamin Franklin, 12203 Berlin, Germany
| | - Uta Demel
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité (Junior) (Digital) Clinician Scientist Program, 10178 Berlin, Germany
| | - Anja A Kühl
- iPATH.Berlin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Benjamin Franklin, 12203 Berlin, Germany
| | - Tim Wartewig
- Institute for Clinical Chemistry and Pathobiochemistry, Technische Universität München, 81675 Munich, Germany
- Center of Molecular and Cellular Oncology, Yale School of Medicine, Yale University, New Haven, CT 06510, USA
| | - Jürgen Ruland
- Institute for Clinical Chemistry and Pathobiochemistry, Technische Universität München, 81675 Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and Technische Universität München, 81675 Munich, Germany
| | - Frank T Wunderlich
- Obesity and Cancer, Max Planck Institute for Metabolism Research, 50931 Cologne, Germany
| | - Markus Schick
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
- German Cancer Consortium (DKTK), partner site Berlin, a partnership between DKFZ and Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Wolfgang Walther
- Experimental and Clinical Research Center, Charité Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Robert-Rössle Str. 10, 13125 Berlin, Germany
- EPO GmbH Berlin-Buch, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-Universität zu Kiel, 24118 Kiel, Germany
| | - Simon Haas
- German Cancer Consortium (DKTK), partner site Berlin, a partnership between DKFZ and Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 10115 Berlin, Germany
- Division of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ) and DKFZ - ZMBH Alliance, 69120 Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany
| | - Leticia Quintanilla-Martinez
- Institute of Pathology and Neuropathology, Comprehensive Cancer Center, Eberhard Karls University of Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," Eberhard Karls University, 72076 Tübingen, Germany
- German Cancer Consortium (DKTK), partner site Tübingen, a partnership between DKFZ and Eberhard Karls University of Tübingen, 72076 Tübingen, Germany
| | - Stefan Feske
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Stephan Ehl
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, University Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Rainer Glauben
- German Cancer Consortium (DKTK), partner site Berlin, a partnership between DKFZ and Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Ulrich Keller
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
- German Cancer Consortium (DKTK), partner site Berlin, a partnership between DKFZ and Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| |
Collapse
|
7
|
Yao R, Huo AP, Jia Y, Su Y. Psoriatic arthritis: Overcoming the obstacles of early diagnosis and optimal management. Int J Rheum Dis 2024; 27:e15015. [PMID: 38287560 DOI: 10.1111/1756-185x.15015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/09/2023] [Accepted: 12/13/2023] [Indexed: 01/31/2024]
Affiliation(s)
- Ranran Yao
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - An-Ping Huo
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yuan Jia
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Yin Su
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
- Peking University People's Hospital, Qingdao, China
| |
Collapse
|
8
|
Cozzi G, Scagnellato L, Lorenzin M, Savarino E, Zingone F, Ometto F, Favero M, Doria A, Vavricka SR, Ramonda R. Spondyloarthritis with inflammatory bowel disease: the latest on biologic and targeted therapies. Nat Rev Rheumatol 2023:10.1038/s41584-023-00984-8. [PMID: 37386288 DOI: 10.1038/s41584-023-00984-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2023] [Indexed: 07/01/2023]
Abstract
Spondyloarthritis (SpA) encompasses a heterogeneous group of chronic inflammatory diseases that can affect both axial and peripheral joints, tendons and entheses. Among the extra-articular manifestations, inflammatory bowel disease (IBD) is associated with considerable morbidity and effects on quality of life. In everyday clinical practice, treatment of these conditions requires a close collaboration between gastroenterologists and rheumatologists to enable early detection of joint and intestinal manifestations during follow-up and to choose the most effective therapeutic regimen, implementing precision medicine for each patient's subtype of SpA and IBD. The biggest issue in this field is the dearth of drugs that are approved for both diseases, as only TNF inhibitors are currently approved for the treatment of full-spectrum SpA-IBD. Janus tyrosine kinase inhibitors are among the most promising drugs for the treatment of peripheral and axial SpA, as well as for intestinal manifestations. Other therapies such as inhibitors of IL-23 and IL-17, phosphodiesterase 4 inhibitor, α4β7 integrin blockers and faecal microbiota transplantation seem to only be able to control some disease domains, or require further studies. Given the growing interest in the development of novel drugs to treat both conditions, it is important to understand the current state of the art and the unmet needs in the management of SpA-IBD.
Collapse
Affiliation(s)
- Giacomo Cozzi
- Rheumatology Unit, Department of Medicine-DIMED, Padova University Hospital, Padova, Italy
| | - Laura Scagnellato
- Rheumatology Unit, Department of Medicine-DIMED, Padova University Hospital, Padova, Italy
| | - Mariagrazia Lorenzin
- Rheumatology Unit, Department of Medicine-DIMED, Padova University Hospital, Padova, Italy
| | - Edoardo Savarino
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University Hospital of Padova, Padova, Italy
| | - Fabiana Zingone
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University Hospital of Padova, Padova, Italy
| | - Francesca Ometto
- Rheumatology Unit, Department of Medicine-DIMED, Padova University Hospital, Padova, Italy
| | - Marta Favero
- Rheumatology Unit, Department of Medicine-DIMED, Padova University Hospital, Padova, Italy
| | - Andrea Doria
- Rheumatology Unit, Department of Medicine-DIMED, Padova University Hospital, Padova, Italy
| | - Stephan R Vavricka
- Department of Gastroenterology and Hepatology, University Hospital Zürich and Center for Gastroenterology and Hepatology, Zürich, Switzerland
| | - Roberta Ramonda
- Rheumatology Unit, Department of Medicine-DIMED, Padova University Hospital, Padova, Italy.
| |
Collapse
|
9
|
Bai LK, Su YZ, Ning ZD, Zhang CQ, Zhang LY, Zhang GL. Challenges and opportunities in animal models of psoriatic arthritis. Inflamm Res 2023:10.1007/s00011-023-01752-w. [PMID: 37300584 DOI: 10.1007/s00011-023-01752-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
OBJECTIVE To review the preparation, characteristics and research progress of different PsA animal models. METHODS Computerized searches were conducted in CNKI, PubMed and other databases to classify and discuss the relevant studies on PsA animal models. The search keywords were "PsA and animal model(s), PsA and animal(s), PsA and mouse, PsA and mice, PsA and rat(s), PsA and rabbit(s), PsA and dog(s)" RESULTS: The experimental animals currently used to study PsA are mainly rodents, including mice and rats. According to the different methods of preparing the models, the retrieved animal models were classified into spontaneous or genetic mutation, transgenic and induced animal models. These PsA animal models involve multiple pathogenesis, some experimental animals' lesions appear in a short and comprehensive cycle, some have a high success rate in molding, and some are complex and less reproducibility. This article summarizes the preparation methods, advantages and disadvantages of different models. CONCLUSIONS The animal models of PsA aim to mimic the clinicopathological alterations of PsA patients through gene mutation, transgenesis or targeted proinflammatory factor and to reveal new pathogenic pathways and therapeutic targets by exploring the pathological features and clinical manifestations of the disease. This work will have very far-reaching implications for the in-depth understanding of PsA and the development of new drugs.
Collapse
Affiliation(s)
- Lin-Kun Bai
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, Shanxi, China
| | - Ya-Zhen Su
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, Shanxi, China
| | - Zong-Di Ning
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, Shanxi, China
| | - Cheng-Qiang Zhang
- Fifth Hospital of Shanxi Medical University, Shanxi Provincial People's Hospital, Taiyuan, 030012, Shanxi, China
| | - Li-Yun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, Shanxi, China
| | - Gai-Lian Zhang
- Fifth Hospital of Shanxi Medical University, Shanxi Provincial People's Hospital, Taiyuan, 030012, Shanxi, China.
| |
Collapse
|
10
|
Niu M, Yuan J, Yan M, Yang G, Yan Z, Yang X. Discovery of CLEC2B as a diagnostic biomarker and screening of celastrol as a candidate drug for psoriatic arthritis through bioinformatics analysis. J Orthop Surg Res 2023; 18:390. [PMID: 37246213 DOI: 10.1186/s13018-023-03843-0] [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] [Received: 04/10/2023] [Accepted: 05/08/2023] [Indexed: 05/30/2023] Open
Abstract
BACKGROUND Psoriatic arthritis (PSA) is a chronic, immune-mediated inflammatory joint disease that is liked to mortality due to cardiovascular disease. Diagnostic markers and effective therapeutic options for PSA remain limited due to the lack of understanding of the pathogenesis. We aimed to identify potential diagnostic markers and screen the therapeutic compounds for PSA based on bioinformatics analysis. METHODS Differentially expressed genes (DEGs) of PSA were identified from the GSE61281 dataset. WGCNA was used to identify PSA-related modules and prognostic biomarkers. Clinical samples were collected to validate the expression of the diagnostic gene. These DEGs were subjected to the CMap database for the identification of therapeutic candidates for PSA. Potential pathways and targets for drug candidates to treat PSA were predicted using Network Pharmacology. Molecular docking techniques were used to validate key targets. RESULTS CLEC2B was identified as a diagnostic marker for PSA patients (AUC > 0.8) and was significantly upregulated in blood samples. In addition, celastrol was identified as a candidate drug for PSA. Subsequently, the network pharmacology approach identified four core targets (IL6, TNF, GAPDH, and AKT1) of celastrol and revealed that celastrol could treat PSA by modulating inflammatory-related pathways. Finally, molecular docking demonstrated stable binding of celastrol to four core targets in the treatment of PSA. Animal experiments indicated celastrol alleviated inflammatory response in the mannan-induced PSA. CONCLUSION CLEC2B was a diagnostic marker for PSA patients. Celastrol was identified as a potential therapeutic drug for PSA via regulating immunity and inflammation.
Collapse
Affiliation(s)
- Min Niu
- Department of Rheumatology Immunology and Endocrinology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Jingman Yuan
- Department of Rheumatology Immunology and Endocrinology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Meixi Yan
- Department of Rheumatology Immunology and Endocrinology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Ge Yang
- Department of Rheumatology Immunology and Endocrinology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Ziyi Yan
- Department of Rheumatology Immunology and Endocrinology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Xichao Yang
- Department of Rheumatology Immunology and Endocrinology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
11
|
Li M, Zhang B, Yin Y, Wen J, Wang J, He Y, Jiang Q, Loor JJ, Wang S, Yang W, Xu C. Intracellular Ca 2+ Signaling and Calcium Release-Activated Calcium Modulator ORAI1 Are Associated With CD4 + T Lymphocytes in Dairy Cows. Front Immunol 2022; 13:835936. [PMID: 35663942 PMCID: PMC9159785 DOI: 10.3389/fimmu.2022.835936] [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: 12/24/2021] [Accepted: 04/19/2022] [Indexed: 11/17/2022] Open
Abstract
The nutritional status of dairy cows and the metabolism of specific nutrients are critical regulators of immune cell function. Around the time of parturition, mobilization of body lipid and muscle helps compensate for the decrease in nutrient intake and the increased requirements of the mammary gland for lactation. An end-result of these processes is the marked increase in circulating concentrations of fatty acids (FA), which are a major risk factor for immune dysfunction. In food animal species such as dairy cows, any disturbance in nutritional or immunological homeostasis leads to deleterious feedback loops that can further risk health, efficiency of nutrient use, and compromise availability of safe and nutritious dairy foods for humans. Despite substantial progress with respect to regulation of innate immunity, such knowledge for adaptive immunity is scarce. To help bridge this gap in knowledge, we sought to study the role of calcium release-activated calcium modulator ORAI1 activation in T cells systemic immune function in vivo. CD4+ T cells were isolated from peripheral blood of dairy cows diagnosed as healthy or with ketosis, a common metabolic disorder of FA metabolism. Results revealed that levels of intracellular Ca2+ and reactive oxygen species (ROS) along with the abundance of store-operated Ca2+ entry (SOCE) moiety increased during ketosis. Further, plasma concentrations of inflammatory cytokines were elevated, the balance of Th17/Treg cells was disrupted, mitochondrial function impaired, and the abundance of mitophagy-related proteins in CD4+ T cells altered during ketosis. Molecular characterization of the direct effects of FA was evaluated in CD4+ T cells isolated from the spleen of 1-day-old calves. Enhanced supply of FA increased intracellular Ca2+ and ROS concentrations, upregulated the abundance of proteins associated with mitochondrial dynamics and ORAI1. Intermediates of mitophagy accumulated and the balance of Th17/Treg cells also was affected by the supply of FA. These negative effects were attenuated by silencing or inhibition of ORAI1 in CD4+ T cells. Together, data indicated that physiological states that lead to increases in systemic concentrations of FA could impact adaptive immunity negatively through ORAI1 regulated intracellular Ca2+, ROS balance, and increased effector functions of Th17 cells.
Collapse
Affiliation(s)
- Ming Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Bingbing Zhang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Yufeng Yin
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Jianan Wen
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Jingjing Wang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Yuxin He
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Qianming Jiang
- Mammalian NutriPhysio Genomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL, United States
| | - Juan J Loor
- Mammalian NutriPhysio Genomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL, United States
| | - Shuang Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Wei Yang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Chuang Xu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| |
Collapse
|
12
|
Macaubas C, Rahman SS, Lavi I, Haddad A, Elias M, Sengupta D, Zisman D, Mellins ED. High Dimensional Analyses of Circulating Immune Cells in Psoriatic Arthritis Detects Elevated Phosphorylated STAT3. Front Immunol 2022; 12:758418. [PMID: 35087513 PMCID: PMC8787828 DOI: 10.3389/fimmu.2021.758418] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 12/20/2021] [Indexed: 01/31/2023] Open
Abstract
Psoriatic arthritis (PsA) is a chronic inflammatory arthritis, affecting up to 40% of patients with psoriasis. Constitutive expression by CD4+ T cells of an active form of STAT3, a signal transducer and transcription factor, has been shown to induce many of the major features of PsA in an animal model. We used high dimensional mass cytometry (CyTOF) to probe ex-vivo levels of phosphorylated STAT3 (pSTAT3) in circulating immune cell subpopulations from PsA patients during active and inactive states. We evaluated the frequency of 16 immune cell populations and the levels of the activated forms of STAT3 (pSTAT3) and, for comparison, STAT1 (pSTAT1) and Src (pSrc) in whole blood fixed shortly after collection. In addition to PsA patients, we studied active rheumatoid arthritis (RA) patients. Increased levels of pSTAT3 were found in all the CD4+ T cell subsets analyzed, specifically, Th1, Th2, Th17, T follicular helper (Tfh) and T regulatory (Treg) as well as in CD14+CD16- (classical) monocytes from active PsA patients compared to inactive patients. After correcting for body mass index (BMI), smoking and conventional disease modifying antirheumatic drugs (c-DMARDs), levels of pSTAT3 levels remained increased in Th1 and Tfh CD4+ T cells, and in CD14+CD16- monocytes from active patients compared to inactive patients. No differences between the patient groups were observed for pSTAT1 or pSrc. No differences were found between the active PsA and active RA groups after correction for multiple testing. During active PsA, circulating Th1 and Tfh CD4+ T cells, and CD14+CD16- monocytes expressing high levels of pSTAT3 may play a role in PsA pathophysiology, perhaps by migration to inflamed sites.
Collapse
Affiliation(s)
- Claudia Macaubas
- Pediatrics, Program in Immunology, Stanford University, Stanford, CA, United States
| | - Shamma S Rahman
- Pediatrics, Program in Immunology, Stanford University, Stanford, CA, United States
| | - Idit Lavi
- Community Medicine and Epidemiology, Carmel Medical Center, Haifa, Israel
| | - Amir Haddad
- Rheumatology Unit, Carmel Medical Center, Haifa, Israel
| | - Muna Elias
- Rheumatology Unit, Carmel Medical Center, Haifa, Israel
| | | | - Devy Zisman
- Rheumatology Unit, Carmel Medical Center, Haifa, Israel.,The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Elizabeth D Mellins
- Pediatrics, Program in Immunology, Stanford University, Stanford, CA, United States
| |
Collapse
|
13
|
Batko B. Exploring the Diverse Immune and Genetic Landscape of Psoriatic Arthritis. J Clin Med 2021; 10:jcm10245926. [PMID: 34945224 PMCID: PMC8706996 DOI: 10.3390/jcm10245926] [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: 11/16/2021] [Revised: 12/12/2021] [Accepted: 12/14/2021] [Indexed: 11/16/2022] Open
Abstract
Psoriatic arthritis (PsA) is characterized by delays in diagnosis and modest effect of treatment in terms of joint response. An understanding of molecular pathomechanisms may aid in developing diagnostic and prognostic models. Genetic susceptibility (e.g., HLA class I genes, IL-23-related genes) can be responsible for the pattern of psoriatic manifestations and affinity for tissue involvement. Gene expression analysis indicates an inflammatory profile that is distinct for PsA, but disparate across tissues. This has clinical implications, as for example, dual blockade of IL-17A and IL-17F can lead to superior clinical effects if there is differential expression of IL-17 receptors in tissues. Structural and functional impairment of barrier tissue, including host-microbiome interactions, may be the source of immune activation. Interplay between different cell populations of innate and adaptive immunity is emerging, potentially providing a link between the transition of skin-to-joint disease. Th17 subsets, IL-17A, IL-17F and IL-23 are crucial in PsA pathogenesis, with both clinical and experimental evidence suggesting a differential molecular landscape in cutaneous and articular compartments.
Collapse
Affiliation(s)
- Bogdan Batko
- Department of Rheumatology and Immunology, Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski University, 30-705 Krakow, Poland
| |
Collapse
|
14
|
Bolt JW, van Ansenwoude CMJ, Hammoura I, van de Sande MG, van Baarsen LGM. Translational Research Studies Unraveling the Origins of Psoriatic Arthritis: Moving Beyond Skin and Joints. Front Med (Lausanne) 2021; 8:711823. [PMID: 34485340 PMCID: PMC8415974 DOI: 10.3389/fmed.2021.711823] [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: 05/19/2021] [Accepted: 07/12/2021] [Indexed: 11/13/2022] Open
Abstract
Patients with psoriatic arthritis (PsA) are suffering from a decreased quality of life despite currently available treatments. In the latest years, novel therapies targeting the IL-17/IL-23 and TNF pathways improved clinical outcome. Despite this, remission of disease is not achieved in a considerable group of patients, continuous treatment is very often required to reach clinical remission, and prevention of PsA in patients with psoriasis (PsO) is currently impossible. A better understanding of PsA pathogenesis is required to develop novel treatment strategies that target inflammation and destruction more effectively and at an early stage of the disease, or even before clinically manifest disease. The skin is considered as one of the sites of onset of immune activation, triggering the inflammatory cascade in PsA. PsO develops into PsA in 30% of the PsO patients. Influenced by environmental and genetic factors, the inflammatory process in the skin, entheses, and/or gut may evolve into synovial tissue inflammation, characterized by influx of immune cells. The exact role of the innate and adaptive immune cells in disease pathogenesis is not completely known. The involvement of activated IL-17A+ T cells could implicate early immunomodulatory events generated in lymphoid organs thereby shaping the pathogenic inflammatory response leading to disease. In this perspective article, we provide the reader with an overview of the current literature regarding the immunological changes observed during the earliest stages of PsA. Moreover, we will postulate future areas of translational research aimed at increasing our knowledge on the molecular mechanisms driving disease development, which will aid the identification of novel potential therapeutic targets to limit the progression of PsA.
Collapse
Affiliation(s)
- Janne W. Bolt
- Department of Rheumatology & Clinical Immunology, Amsterdam Institute for Infection & Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Immunology, Amsterdam Institute for Infection & Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Rheumatology & Immunology Center (ARC), Academic Medical Center, Amsterdam, Netherlands
| | - Chaja M. J. van Ansenwoude
- Department of Rheumatology & Clinical Immunology, Amsterdam Institute for Infection & Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Immunology, Amsterdam Institute for Infection & Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Rheumatology & Immunology Center (ARC), Academic Medical Center, Amsterdam, Netherlands
| | - Ihsan Hammoura
- Department of Rheumatology & Clinical Immunology, Amsterdam Institute for Infection & Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Immunology, Amsterdam Institute for Infection & Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Rheumatology & Immunology Center (ARC), Academic Medical Center, Amsterdam, Netherlands
| | - Marleen G. van de Sande
- Department of Rheumatology & Clinical Immunology, Amsterdam Institute for Infection & Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Immunology, Amsterdam Institute for Infection & Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Rheumatology & Immunology Center (ARC), Academic Medical Center, Amsterdam, Netherlands
| | - Lisa G. M. van Baarsen
- Department of Rheumatology & Clinical Immunology, Amsterdam Institute for Infection & Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Immunology, Amsterdam Institute for Infection & Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Rheumatology & Immunology Center (ARC), Academic Medical Center, Amsterdam, Netherlands
| |
Collapse
|
15
|
Mandour M, Chen S, van de Sande MGH. The Role of the IL-23/IL-17 Axis in Disease Initiation in Spondyloarthritis: Lessons Learned From Animal Models. Front Immunol 2021; 12:618581. [PMID: 34267743 PMCID: PMC8276000 DOI: 10.3389/fimmu.2021.618581] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 05/31/2021] [Indexed: 12/14/2022] Open
Abstract
Spondyloarthritis (SpA) is a spectrum of chronic inflammatory joint diseases that frequently presents with inflammation of the axial skeleton, peripheral joints, entheses, skin, and gut. Understanding SpA pathogenesis has been proven challenging due to the limited availability of human target tissues. In recent years, the interleukin (IL)-23/IL-17 pathway has been implicated in the pathogenesis of SpA, in addition to the Tumor Necrosis Factor Alpha (TNF-α) cytokine. The underlying molecular mechanisms by which the IL-23/IL-17 pathway triggers disease initiation, both in the joints as well as at extra-musculoskeletal sites, are not precisely known. Animal models that resemble pathological features of human SpA have provided possibilities for in-depth molecular analyses of target tissues during various phases of the disease, including the pre-clinical initiation phase of the disease before arthritis and spondylitis are clinically present. Herein, we summarize recent insights gained in SpA animal models on the role of the IL-23/IL-17 pathway in immune activation across affected sites in SpA, which include the joint, entheses, gut and skin. We discuss how local activation of the IL-23/IL-17 axis may contribute to the development of tissue inflammation and the onset of clinically manifest SpA. The overall aim is to provide the reader with an overview of how the IL-23/IL-17 axis could contribute to the onset of SpA pathogenesis. We discuss how insights from animal studies into the initiation phase of disease could instruct validation studies in at-risk individuals and thereby provide a perspective for potential future preventive treatment.
Collapse
Affiliation(s)
- Mohamed Mandour
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Immunology, Infection and Immunity Institute, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Sijia Chen
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Immunology, Infection and Immunity Institute, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam, Netherlands
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Marleen G. H. van de Sande
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Immunology, Infection and Immunity Institute, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
16
|
Ceribelli A, Motta F, Vecellio M, Isailovic N, Ciccia F, Selmi C. Clinical Trials Supporting the Role of the IL-17/IL-23 Axis in Axial Spondyloarthritis. Front Immunol 2021; 12:622770. [PMID: 34149686 PMCID: PMC8206811 DOI: 10.3389/fimmu.2021.622770] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 05/14/2021] [Indexed: 12/21/2022] Open
Abstract
The term spondyloarthritis (SpA) encompasses a heterogeneous group of inflammatory musculoskeletal diseases with several common genetic background and clinical features, including the possible involvement of the axial skeleton with peripheral mono- or oligo- arthritis and frequently coexisting skin, eye and intestinal manifestations. When the sacroiliac joints or other parts of the spine or thoracic wall are predominantly affected at magnetic resonance or X-ray imaging with inflammatory back pain, the disease is classified as axial SpA and the therapeutic choices are significantly different compared to cases of peripheral arthritis. Moving from the narrow effectiveness and safety profiles of non-steroidal anti-inflammatory drugs, there has been a significant research effort aimed at identifying new treatments based on our better understanding of the pathogenesis of SpA. Indeed, in parallel with the solid data demonstrating that IL-17 and IL-23 are key cytokines in the development of enthesitis and spondylitis, monoclonal antibodies interfering with this pathway have been developed for the treatment of axial SpA. Furthermore, the IL-17/IL-23 axis is key to extra-articular manifestations such as inflammatory bowel disease, uveitis, and psoriasis which are frequent comorbidities of SpA. Currently available drugs act through these mechanisms recognizing IL-23 and targeting IL-17, such as secukinumab and ixekizumab. These therapeutic approaches are now envisioned in the international treatment recommendations for psoriatic arthritis with an axial phenotype as well as for ankylosing spondylitis (AS). We will provide herein a concise comprehensive overview of the clinical evidence supporting the use of these and other drugs acting on IL-23 and IL-17 in axial SpA.
Collapse
Affiliation(s)
- Angela Ceribelli
- Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center - IRCCS, Rozzano (Mi), Italy
- Department of Biomedical Sciences, Humanitas University, Rozzano (Mi), Italy
| | - Francesca Motta
- Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center - IRCCS, Rozzano (Mi), Italy
- Department of Biomedical Sciences, Humanitas University, Rozzano (Mi), 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, Oxford, United Kingdom
| | - Natasa Isailovic
- Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center - IRCCS, Rozzano (Mi), Italy
| | - Francesco Ciccia
- Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Carlo Selmi
- Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center - IRCCS, Rozzano (Mi), Italy
- Department of Biomedical Sciences, Humanitas University, Rozzano (Mi), Italy
| |
Collapse
|
17
|
Al-Bogami M, Bystrom J, Clanchy F, Taher TE, Mangat P, Williams RO, Jawad AS, Mageed RA. TNFα inhibitors reduce bone loss in rheumatoid arthritis independent of clinical response by reducing osteoclast precursors and IL-20. Rheumatology (Oxford) 2021; 60:947-957. [PMID: 32984900 DOI: 10.1093/rheumatology/keaa551] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/17/2020] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES About half of RA patients treated with TNFα inhibitors either do not respond or lose their initial therapeutic response over time. The clinical response is measured by reduction in DAS28, which primarily reflects inflammation. However, other effects of TNFα inhibitors, such as impact on bone erosion, are not assessed by DAS28. We aimed to examine the effect of TNFα inhibitors on bone density, bone biomarkers and cytokine production in responder and non-responder patients and assessed mechanisms of action. METHODS BMD in the lumbar spine and femur neck of 117 RA patients was measured by DEXA scan. Bone turnover biomarkers CTX, osteoprotegerin (OPG), osteocalcin and RANKL were measured by ELISA. Levels of 16 cytokines in plasma and in tissue culture supernatants of ex vivo T cells were measured by multiplex assays and ELISA. The effect of treatment with TNFα inhibitors on blood mononuclear cell (MNC) differentiation to osteoclast precursors (OCP) was measured flow cytometry and microscopy. RESULTS TNFα inhibitors improved lumbar spine BMD but had modest effects on blood bone biomarkers, irrespective of patients' clinical response. Blood OCP numbers and the ability of monocytes to differentiate to OCP in vitro declined after treatment. Treatment also reduced RANK expression and IL-20 production. BMD improvement correlated with reduced levels of IL-20 in responder patients. CONCLUSION This study reveals that TNFα inhibitors reduce lumbar spine bone loss in RA patients irrespective of changes in DAS28. The reduction in bone loss is associated with reduction in IL-20 levels in responder patients.
Collapse
Affiliation(s)
- Mohammed Al-Bogami
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Jonas Bystrom
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Felix Clanchy
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Taher E Taher
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Pamela Mangat
- Department of Rheumatology, Royal Free Hospital, NHS Foundation Trust London, London, UK
| | | | - Ali S Jawad
- Department of Rheumatology, Barts Health NHS Trust, London, UK
| | - Rizgar A Mageed
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK
| |
Collapse
|
18
|
Yang K, Oak ASW, Elewski BE. Use of IL-23 Inhibitors for the Treatment of Plaque Psoriasis and Psoriatic Arthritis: A Comprehensive Review. Am J Clin Dermatol 2021; 22:173-192. [PMID: 33301128 PMCID: PMC7727454 DOI: 10.1007/s40257-020-00578-0] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Psoriasis is a common inflammatory skin disease with multiple comorbidities, including psoriatic arthritis and coronary artery disease, that can severely impact an individual’s quality of life and daily functioning. In recent years, enhanced understanding of the pathogenesis of psoriasis, especially the role of T helper 17 cells, has resulted in the development of new classes of biologic drugs targeting modulators along its disease pathway. Among these, inhibitors of interleukin-23 (e.g., ustekinumab, guselkumab, tildrakizumab, and risankizumab) have emerged as safe and effective options for the treatment of moderate-to-severe plaque psoriasis; ustekinumab and guselkumab have additionally been approved to treat psoriatic arthritis. Selective interleukin-23 inhibitors require less frequent dosing than interleukin-17 inhibitors and may possess a more favorable risk profile without an increased risk of candidiasis or inflammatory bowel disease. Overall, these highly effective medications are contributing to a rising standard for psoriasis outcomes through resolution of skin lesions and joint manifestations and improvement of patient quality of life.
Collapse
|
19
|
Abstract
Interleukin-22 (IL-22) is secreted by a wide range of immune cells and its downstream effects are mediated by the IL-22 receptor, which is present on non-immune cells in many organs throughout the body. IL-22 is an inflammatory mediator that conditions the tissue compartment by upregulating innate immune responses and is also a homeostatic factor that promotes tissue integrity and regeneration. Interestingly, the IL-22 system has also been linked to many T cell driven inflammatory diseases. Despite this, the downstream effects of IL-22 on the adaptive immune system has received little attention. We have reviewed the literature for experimental data that suggest IL-22 mediated effects on T cells, either transduced directly or via mediators expressed by innate immune cells or non-immune cells in response to IL-22. Collectively, the reviewed data indicate that IL-22 has a hitherto unappreciated influence on T helper cell polarization, or the secretion of signature cytokines, that is context dependent but in many cases results in a reduction of the Th1 type response and to some extent promotion of regulatory T cells. Further studies are needed that specifically address these aspects of IL-22 signaling, which can benefit the understanding and treatment of a wide range of diseases.
Collapse
Affiliation(s)
- Hannes Lindahl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
20
|
Yang KL, Lejeune A, Chang G, Scher JU, Koralov SB. Microbial-derived antigens and metabolites in spondyloarthritis. Semin Immunopathol 2021; 43:163-172. [PMID: 33569635 DOI: 10.1007/s00281-021-00844-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 01/20/2021] [Indexed: 12/30/2022]
Abstract
Spondyloarthritis (SpA) is a group of chronic, immune-mediated, inflammatory diseases affecting the bone, synovium, and enthesis. Microbiome, the community of microorganisms that has co-evolved with human hosts, plays a pivotal role in human health and disease. This invisible "essential organ" supplies the host with a myriad of chemicals and molecules. In turn, microbial metabolites can serve as messengers for microbes to communicate with each other and in the cross-talk with host cells. Gut dysbiosis in SpA is associated with altered microbial metabolites, and an accumulated body of research has contributed to the understanding that changes in intestinal microbiota can modulate disease pathogenesis. We review the novel findings from human and animal studies to provide an overview of the contribution of individual microbial metabolites and antigens to SpA.
Collapse
Affiliation(s)
- Katharine Lu Yang
- Department of Pathology, NYU School of Medicine, 522 First Ave. Smilow Research Bldg 511, New York, NY, 10016, USA
| | - Alannah Lejeune
- Department of Pathology, NYU School of Medicine, 522 First Ave. Smilow Research Bldg 511, New York, NY, 10016, USA
| | - Gregory Chang
- Department of Radiology, NYU School of Medicine, New York, NY, 10016, USA
| | - Jose U Scher
- Division of Rheumatology, Department of Medicine, NYU School of Medicine, New York, NY, 10016, USA. .,Division of Rheumatology and Psoriatic Arthritis Center, 301 East 17th St, Room 1608, New York, NY, 10003, USA.
| | - Sergei B Koralov
- Department of Pathology, NYU School of Medicine, 522 First Ave. Smilow Research Bldg 511, New York, NY, 10016, USA.
| |
Collapse
|
21
|
Murray K, Floudas A, Murray C, Fabre A, Crown J, Fearon U, Veale D. First use of tofacitinib to treat an immune checkpoint inhibitor-induced arthritis. BMJ Case Rep 2021; 14:14/2/e238851. [PMID: 33541985 PMCID: PMC7868229 DOI: 10.1136/bcr-2020-238851] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Immune checkpoint inhibitors have revolutionised cancer treatment; however, immune-related adverse events do occur, with up to 7% developing inflammatory arthritis. Common rheumatoid arthritis therapies such as methotrexate, prednisolone and biologics have been used to treat this arthritis in small, uncontrolled case series with varying success. In this case of personalised medicine, we report the first use of tofacitinib, a small molecular inhibitor of the Janus kinase-signal transducer and activator of transcription pathway, to treat checkpoint inhibitor-related inflammatory arthritis. This resulted in a rapid clinical response and complete, sustained remission of the arthritis with associated marked reduction in synovial molecular and cellular immune response.
Collapse
Affiliation(s)
- Kieran Murray
- EULAR Centre for Arthritis and Rheumatic Disease, St. Vincent's University Hospital, Dublin, Ireland
| | - Achilleas Floudas
- EULAR Centre for Arthritis and Rheumatic Disease, St. Vincent's University Hospital, Dublin, Ireland,Molecular Rheumatology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Ciara Murray
- Pathology, Oncology and Medicine, University College Dublin, Dublin, Ireland
| | - Aurelie Fabre
- Pathology, Oncology and Medicine, University College Dublin, Dublin, Ireland
| | - John Crown
- Pathology, Oncology and Medicine, University College Dublin, Dublin, Ireland
| | - Ursula Fearon
- EULAR Centre for Arthritis and Rheumatic Disease, St. Vincent's University Hospital, Dublin, Ireland,Molecular Rheumatology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Douglas Veale
- EULAR Centre for Arthritis and Rheumatic Disease, St. Vincent's University Hospital, Dublin, Ireland,Pathology, Oncology and Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
22
|
Billi AC, Ludwig JE, Fritz Y, Rozic R, Swindell WR, Tsoi LC, Gruzska D, Abdollahi-Roodsaz S, Xing X, Diaconu D, Uppala R, Camhi MI, Klenotic PA, Sarkar MK, Husni ME, Scher JU, McDonald C, Kahlenberg JM, Midura RJ, Gudjonsson JE, Ward NL. KLK6 expression in skin induces PAR1-mediated psoriasiform dermatitis and inflammatory joint disease. J Clin Invest 2021; 130:3151-3157. [PMID: 32155135 DOI: 10.1172/jci133159] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 02/27/2020] [Indexed: 12/12/2022] Open
Abstract
Kallikrein-related peptidase 6 (KLK6) is a secreted serine protease hypothesized to promote inflammation via cleavage of protease-activated receptor 1 (PAR1) and PAR2. KLK6 levels are elevated in multiple inflammatory and autoimmune conditions, but no definitive role in pathogenesis has been established. Here, we show that skin-targeted overexpression of KLK6 causes generalized, severe psoriasiform dermatitis with spontaneous development of debilitating psoriatic arthritis-like joint disease. The psoriatic skin and joint phenotypes are reversed by normalization of skin KLK6 levels and attenuated following genetic elimination of PAR1 but not PAR2. Conservation of this regulatory pathway was confirmed in human psoriasis using vorapaxar, an FDA-approved PAR1 antagonist, on explanted lesional skin from patients with psoriasis. Beyond defining a critical role for KLK6/PAR1 signaling in promoting psoriasis, our results demonstrate that KLK6/PAR1-mediated inflammation in the skin alone is sufficient to drive inflammatory joint disease. Further, we identify PAR1 as a promising cytokine-independent target in therapy of psoriasis and psoriatic arthritis.
Collapse
Affiliation(s)
- Allison C Billi
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jessica E Ludwig
- Department of Dermatology and.,Department of Nutrition, Case Western Reserve University, Cleveland, Ohio, USA
| | | | - Richard Rozic
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - William R Swindell
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA.,Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA.,Department of Internal Medicine, Jewish Hospital, Cincinnati, Ohio, USA
| | - Lam C Tsoi
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA.,Center for Statistical Genetics, Department of Biostatistics, and.,Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - Dennis Gruzska
- Department of Dermatology and.,Department of Nutrition, Case Western Reserve University, Cleveland, Ohio, USA
| | - Shahla Abdollahi-Roodsaz
- Inflammation and Immunology Thematic Center of Excellence, Celgene Corp., Cambridge, Massachusetts, USA
| | - Xianying Xing
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Ranjitha Uppala
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | - Mrinal K Sarkar
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | - M Elaine Husni
- Department of Rheumatologic and Immunologic Disease, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jose U Scher
- Department of Medicine, New York University, New York, New York, USA
| | - Christine McDonald
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | | | - Ronald J Midura
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | | | - Nicole L Ward
- Department of Dermatology and.,Department of Nutrition, Case Western Reserve University, Cleveland, Ohio, USA.,Murdough Family Center for Psoriasis, Cleveland, Ohio, USA
| |
Collapse
|
23
|
Abstract
Juvenile spondyloarthritis is a subset of juvenile idiopathic arthritis (JIA) with onset in late childhood and adolescence and a strong association with human leukocyte antigen (HLA) B-27 positivity and familial aggregation that has the potential for axial involvement, potentially leading to ankylosing spondylitis. Current therapy for severe disease relies heavily on tumor necrosis factor inhibitors (TNFi). Treatment paradigms in children largely consist of extrapolation from studies on adults with spondyloarthritis. Additional therapies studied in adults include non-steroidal anti-inflammatory drugs (NSAIDs), blockade of the interleukin-17 (IL-17) and IL-23 axes, blockade of T-cell stimulation, phosphodiesterase (PDE)-4 inhibition, and Janus-activated kinase (JAK) pathway alteration. IL-17 blockade and IL-23 blockade are guideline approved after TNFi failure (and even as an alternative to TNFi) in adults, depending on concomitant inflammatory bowel and skin disease, with JAK and PDE-4 inhibition options following biologic failure. Neither pediatric nor adult guidelines address IL-6 blockade, T-cell co-stimulation blockade, or combination biologic therapy.
Collapse
Affiliation(s)
- John M Bridges
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Matthew L Stoll
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
24
|
Liu Y, Jarjour W, Olsen N, Zheng SG. Traitor or warrior-Treg cells sneaking into the lesions of psoriatic arthritis. Clin Immunol 2020; 215:108425. [PMID: 32305454 DOI: 10.1016/j.clim.2020.108425] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/14/2020] [Accepted: 04/14/2020] [Indexed: 01/01/2023]
Abstract
Regulatory T (Treg) cells have been recognized to maintain immune tolerance, which contributes to prevention of autoimmune diseases. However, recent evidence has demonstrated different characteristics of these cells between those that are in circulation compared to those in various local tissues. In addition, the ability of Treg cells to have plasticity in certain disease settings and in inflammatory lesions has been increasingly recognized. Herein we summarize updated knowledge of Treg biology and discuss the current understanding of tissue-resident Treg cells in psoriatic arthritis (PsA), attempting to provide new insights into precise role of Treg cells in the immune response and as a possible therapeutic intervention in patients with PsA.
Collapse
Affiliation(s)
- Yan Liu
- Institute of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510620, China
| | - Wael Jarjour
- Department of Internal Medicine, The Ohio State University College of Medicine, Wexner Medical Center, Columbus 43210, USA
| | - Nancy Olsen
- Department of Medicine, The Penn State Hershey College of Medicine, Hershey 17031, USA
| | - Song Guo Zheng
- Department of Internal Medicine, The Ohio State University College of Medicine, Wexner Medical Center, Columbus 43210, USA.
| |
Collapse
|
25
|
Sakkas LI, Zafiriou E, Bogdanos DP. Mini Review: New Treatments in Psoriatic Arthritis. Focus on the IL-23/17 Axis. Front Pharmacol 2019; 10:872. [PMID: 31447673 PMCID: PMC6691125 DOI: 10.3389/fphar.2019.00872] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 07/08/2019] [Indexed: 01/22/2023] Open
Abstract
Psoriasis, an inflammatory skin disease, and psoriatic arthritis (PsA), an inflammatory arthritis, share clinical, genetic, and pathogenic factors and may be summed as one disease, the psoriatic disease. Interleukin (IL)-17 plays a major role in the development of both psoriasis and PsA. IL-23 is important in the proliferation and maintenance of IL-17, and therefore, cytokines of the IL-23/IL-17 axis attracted much interest as therapeutic targets in psoriasis and PsA. Therapeutic agents targeting the IL-23/IL-17 axis have been proven to be very effective in psoriasis and PsA, some are already in the therapeutic armamentarium and others are in the development. Some agents, target IL-23 and others IL-17 and include anti-IL-12/IL-23 p40 (ustekinumab, briankizumab), anti-IL-23p19 (guselkumab, tildrakizumab, risankizumab, brazikumab, mirikizumab), anti-IL-17A (secukinumab, ixekizumab), dual anti-IL-17A and anti-IL-17F (bimekizumab), or anti-IL-17 receptor (brodalumab) monoclonal antibodies. Janus tyrosine kinase(JAK) inhibitors also directly affect IL-23 and, thus, IL-17. After the first-generation pan-JAK inhibitors have been shown efficacy (tofacitinib, baricitinib), new-generation selective JAK inhibitors (filgotinib, upadacitinib) are under investigation in psoriasis and PsA.
Collapse
Affiliation(s)
- Lazaros I Sakkas
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Efterpi Zafiriou
- Department of Dermatology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Dimitrios P Bogdanos
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| |
Collapse
|
26
|
McGonagle DG, McInnes IB, Kirkham BW, Sherlock J, Moots R. The role of IL-17A in axial spondyloarthritis and psoriatic arthritis: recent advances and controversies. Ann Rheum Dis 2019; 78:1167-1178. [PMID: 31278139 PMCID: PMC6788885 DOI: 10.1136/annrheumdis-2019-215356] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 05/17/2019] [Accepted: 06/14/2019] [Indexed: 12/19/2022]
Abstract
Although the pathogenic mechanisms underlying axial spondyloarthritis (axSpA) and psoriatic arthritis (PsA) are not fully elucidated, several lines of evidence suggest that immune responses mediated by interleukin 17A (IL-17A) play a pivotal role in both diseases. This is best highlighted by the significant clinical efficacy shown with inhibitors of IL-17A in treating axSpA and PsA. Nevertheless, a number of knowledge gaps exist regarding the role of IL-17A in the pathophysiology of spondyloarthritis in man, including its cellular origin, its precise role in discrete disease processes such enthesitis, bone erosion, and bone formation, and the reasons for the discrepant responses to IL-17A inhibition observed in certain other spondyloarthritis manifestations. In this review, we focus on the latest data from studies investigating the role of IL-17A in ankylosing spondylitis (AS) and PsA that build on existing and emerging scientific knowledge in the field. Key remaining research questions are also highlighted to guide future research.
Collapse
Affiliation(s)
- Dennis G McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.,NIHR Leeds Musculoskeletal Biomedical Research Unit, Chapel Allerton, Leeds Teaching Hospital Trust, Leeds, UK
| | - Iain B McInnes
- Institute of Infection Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Bruce W Kirkham
- Rheumatology Department, Guy's and Saint Thomas' NHS Foundation Trust, London, UK
| | - Jonathan Sherlock
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.,Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Robert Moots
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK .,Department of Academic Rheumatology, Aintree University Hospital, Liverpool, UK
| |
Collapse
|
27
|
Kaufmann U, Kahlfuss S, Yang J, Ivanova E, Koralov SB, Feske S. Calcium Signaling Controls Pathogenic Th17 Cell-Mediated Inflammation by Regulating Mitochondrial Function. Cell Metab 2019; 29:1104-1118.e6. [PMID: 30773462 PMCID: PMC6506368 DOI: 10.1016/j.cmet.2019.01.019] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 11/26/2018] [Accepted: 01/23/2019] [Indexed: 12/24/2022]
Abstract
Pathogenic Th17 cells play important roles in many autoimmune and inflammatory diseases. Their function depends on T cell receptor (TCR) signaling and cytokines that activate signal transducer and activator of transcription 3 (STAT3). TCR engagement activates stromal interaction molecule 1 (STIM1) and calcium (Ca2+) influx through Ca2+-release-activated Ca2+ (CRAC) channels. Here, we show that abolishing STIM1 and Ca2+ influx in T cells expressing a hyperactive form of STAT3 (STAT3C) attenuates pathogenic Th17 cell function and inflammation associated with STAT3C expression. Deletion of STIM1 in pathogenic Th17 cells reduces the expression of genes required for mitochondrial function and oxidative phosphorylation (OXPHOS) but enhances reactive oxygen species (ROS) production. STIM1 deletion or inhibition of OXPHOS is associated with a non-pathogenic Th17 gene expression signature and impaired pathogenic Th17 cell function. Our findings establish Ca2+ influx as a critical regulator of mitochondrial function and oxidative stress in pathogenic Th17 cell-mediated multiorgan inflammation.
Collapse
Affiliation(s)
- Ulrike Kaufmann
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Sascha Kahlfuss
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Jun Yang
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Elitza Ivanova
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Sergei B Koralov
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Stefan Feske
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
28
|
Wade SM, Canavan M, McGarry T, Low C, Wade SC, Mullan RH, Veale DJ, Fearon U. Association of synovial tissue polyfunctional T-cells with DAPSA in psoriatic arthritis. Ann Rheum Dis 2019; 78:350-354. [PMID: 30626658 PMCID: PMC6390025 DOI: 10.1136/annrheumdis-2018-214138] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 11/29/2018] [Accepted: 12/18/2018] [Indexed: 01/23/2023]
Abstract
OBJECTIVE This study examines polyfunctional T-cells in psoriatic arthritis (PsA) synovial tissue and their associations with clinical disease and implications for therapy. METHODS PsA synovial tissue was enzymatically/mechanically digested to generate synovial tissue single cell suspensions. Frequencies of polyfunctional CD4, CD8, T-helper 1 (Th1), Th17 and exTh17 cells, using CD161 as a marker of Th17 plasticity, were determined by flow cytometry in matched PsA synovial tissue and peripheral blood. Synovial T-cell polyfunctionality was assessed in relation to Disease Activity in PSoriatic Arthritis (DAPSA) and in synovial cell suspensions cultured with a current mode of treatment, phosphodiesterase 4 (PDE4) inhibitor. RESULTS PsA synovial tissue infiltrating CD4+ T-cells expressed higher levels of interleukin (IL)-17A, interferon gamma (IFN-γ), GM-CSF and CD161, with parallel enrichment of Th1, Th17 and exTh17 T-helper subsets (all p<0.05). Interestingly, a significant proportion of synovial T-cell subsets were triple-positive for GM-CSF, tumour necrosis factor (-TNF), -IL-17 or IFN-γ compared with matched blood (all p<0.05). Importantly, frequencies of polyfunctional T-cells correlated with DAPSA: Th1-GM-CSF+/TNF+/IFN-γ+ (r=0.7, p<0.01), Th17-GM-CSF+/TNF+/IL-17+ (r=0.6, p<0.057) and exTh17-GM-CSF+/TNF+/IFN-γ+ (r=0.7, p=0.0096), with no associations observed for single cytokine-producing T-cells. Following ex vivo culture of PsA synovial tissue cell suspensions, polyfunctional GM-CSF+TNFα+IL-17A+ or/IFN-γ+-producing T-cells (p<0.05), but not single cytokine-producing T-cells, were inhibited with a PDE4 inhibitor. CONCLUSION These data demonstrate enrichment of polyfunctional T-cells in PsA synovial tissue which were strongly associated with DAPSA and ex vivo therapeutic response.
Collapse
Affiliation(s)
- Sarah M Wade
- Molecular Rheumatology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.,St Vincent's University Hospital, Dublin Academic Health Care and University College Dublin, Dublin 4, Ireland
| | - Mary Canavan
- Molecular Rheumatology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Trudy McGarry
- Molecular Rheumatology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.,St Vincent's University Hospital, Dublin Academic Health Care and University College Dublin, Dublin 4, Ireland
| | - Candice Low
- St Vincent's University Hospital, Dublin Academic Health Care and University College Dublin, Dublin 4, Ireland
| | - Siobhan C Wade
- Molecular Rheumatology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Ronan H Mullan
- Department of Rheumatology, Tallaght University Hospital, Dublin 24, Ireland
| | - Douglas J Veale
- St Vincent's University Hospital, Dublin Academic Health Care and University College Dublin, Dublin 4, Ireland
| | - Ursula Fearon
- Molecular Rheumatology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
29
|
Gravallese EM, Schett G. Effects of the IL-23–IL-17 pathway on bone in spondyloarthritis. Nat Rev Rheumatol 2018; 14:631-640. [DOI: 10.1038/s41584-018-0091-8] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
30
|
Veale DJ, Fearon U. The pathogenesis of psoriatic arthritis. Lancet 2018; 391:2273-2284. [PMID: 29893226 DOI: 10.1016/s0140-6736(18)30830-4] [Citation(s) in RCA: 326] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 03/21/2018] [Accepted: 03/28/2018] [Indexed: 12/15/2022]
Abstract
Psoriatic arthritis is a chronic, immune-mediated, inflammatory arthropathy that presents with inflammation of the joints and entheses, including those of the axial skeleton, and is associated with increased mortality from cardiovascular disease. Diagnosis is primarily based on clinical phenotype because of the diversity of the associated features, which can include skin and nail disease, dactylitis, uveitis, and osteitis. Improved understanding of the pathogenesis of psoriatic arthritis has led to the development of effective biologics and small-molecular drugs targeting specific cytokines and signalling pathways, which can prevent disease progression and improve quality of life. However, at least 40% of patients with psoriatic arthritis have only a partial response or fail to respond to such treatments. Cytokine inhibitors, mainly those specific for tumour necrosis factor and, more recently, the interleukin 23-T-helper-17 cell pathway, have been highly successful in the treatment of disease manifestations in several different tissues, although targeting the interleukin 23-T-helper-17 cell pathway might be more effective in psoriasis than in arthritis. However, the precise mechanisms underlying the pathogenesis of psoriatic arthritis-which include genetics, environmental factors, and immune-mediated inflammation-are complex, and the relationship between disease of the joint and that of other domains is poorly understood. Improving our understanding of psoriatic arthritis pathogenesis could help to establish validated biomarkers for diagnosis, predict therapeutic response and remission, develop precision medicines, and predict which patients will respond to which therapy. We discuss advances in pathogenetic translational research that could inform these issues.
Collapse
Affiliation(s)
- Douglas J Veale
- Rheumatology EULAR Centre of Excellence, St Vincent's University Hospital and University College Dublin, Dublin, Ireland.
| | - Ursula Fearon
- Rheumatology EULAR Centre of Excellence, St Vincent's University Hospital and University College Dublin, Dublin, Ireland; Department of Molecular Rheumatology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
31
|
Mountz JD. Editorial: STATus of STAT3 in Psoriatic Arthritis. Arthritis Rheumatol 2018; 70:801-804. [PMID: 29439293 DOI: 10.1002/art.40445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 02/06/2018] [Indexed: 02/02/2023]
|