1
|
Agarwal A, Maikap D, Padhan P. Treatment of Reactive Arthritis with Biological Agents. Curr Rheumatol Rep 2024; 26:450-458. [PMID: 39312088 DOI: 10.1007/s11926-024-01165-6] [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] [Accepted: 09/04/2024] [Indexed: 11/01/2024]
Abstract
PURPOSE OF THE REVIEW Reactive arthritis (ReA) is an inflammatory joint condition triggered by an infection elsewhere in the body, and this review aims to provide a comprehensive synthesis of recent studies including case reports and case series to determine whether biologics are a treatment option. RECENT FINDINGS Recent studies indicate that biological agents, including anti-TNF agents (infliximab, adalimumab, etanercept), anti-IL17 (secukinumab), and anti-IL6 (tocilizumab), are effective in treating refractory cases of ReA. Evidence suggests these agents are associated with significant clinical improvement. Notably, the data reveal that these biologics are generally well-tolerated, with a low incidence of major adverse events, which supports their safety profile for use in ReA. Biological agents, including anti-TNF, anti-IL17, and anti-IL6 therapies, can be safely and effectively used in the treatment of ReA when conventional therapies fail. It further emphasizes the need for a well-designed controlled trial to provide scientific basis for better informed clinical decisions in cases not responding to conventional treatment.
Collapse
Affiliation(s)
- Avarna Agarwal
- Department of Clinical Immunology and Rheumatology, Kalinga Institute of Medical Sciences, KIIT University, Bhubaneswar, Odisha, India
| | - Debashis Maikap
- Department of Clinical Immunology and Rheumatology, Kalinga Institute of Medical Sciences, KIIT University, Bhubaneswar, Odisha, India
| | - Prasanta Padhan
- Department of Clinical Immunology and Rheumatology, Kalinga Institute of Medical Sciences, KIIT University, Bhubaneswar, Odisha, India.
| |
Collapse
|
2
|
Neurath L, Sticherling M, Schett G, Fagni F. Targeting cytokines in psoriatic arthritis. Cytokine Growth Factor Rev 2024; 78:1-13. [PMID: 39068140 DOI: 10.1016/j.cytogfr.2024.06.001] [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: 06/10/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/30/2024]
Abstract
Psoriatic arthritis (PsA) is part of the psoriatic disease spectrum and is characterized by a chronic inflammatory process that affects entheses, tendons and joints. Cytokines produced by immune and non-immune cells play a central role in the pathogenesis of PsA by orchestrating key aspects of the inflammatory response. Pro-inflammatory cytokines such as TNF, IL-23 and IL-17 have been shown to regulate the initiation and progression of PsA, ultimately leading to the destruction of the architecture of the local tissues such as soft tissue, cartilage and bone. The important role of cytokines in PsA has been underscored by the clinical success of antibodies that neutralize their function. In addition to biologic agents targeting individual pro-inflammatory cytokines, signaling inhibitors that block multiple cytokines simultaneously such as JAK inhibitors have been approved for PsA therapy. In this review, we will focus on our current understanding of the role of cytokines in the disease process of PsA and discuss potential new treatment options based on modulation of cytokine function.
Collapse
Affiliation(s)
- Laura Neurath
- Department of Internal Medicine 3, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie DZI, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Michael Sticherling
- Deutsches Zentrum Immuntherapie DZI, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany; Department of Dermatology, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie DZI, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Filippo Fagni
- Department of Internal Medicine 3, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum Immuntherapie DZI, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany.
| |
Collapse
|
3
|
Hammitzsch A, Ossadnik A, Bachmann Q, Merwald-Fraenk H, Lorenz G, Witt M, Wiesent F, Mühlhofer H, Simone D, Bowness P, Heemann U, Arbogast M, Moog P, Schmaderer C. Increased interleukin-26 in the peripheral joints of patients with axial spondyloarthritis and psoriatic arthritis, co-localizing with CD68-positive synoviocytes. Front Immunol 2024; 15:1355824. [PMID: 38799447 PMCID: PMC11127564 DOI: 10.3389/fimmu.2024.1355824] [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: 12/14/2023] [Accepted: 04/01/2024] [Indexed: 05/29/2024] Open
Abstract
Objectives IL26 levels are elevated in the blood and synovial fluid of patients with inflammatory arthritis. IL26 can be produced by Th17 cells and locally within joints by tissue-resident cells. IL26 induces osteoblast mineralization in vitro. As osteoproliferation and Th17 cells are important factors in the pathogenesis of axial spondyloarthritis (axSpA), we aimed to clarify the cellular sources of IL26 in spondyloarthritis. Methods Serum, peripheral blood mononuclear cells (n = 15-35) and synovial tissue (n = 3-9) of adult patients with axSpA, psoriatic arthritis (PsA) and rheumatoid arthritis (RA) and healthy controls (HCs, n = 5) were evaluated by ELISA, flow cytometry including PrimeFlow assay, immunohistochemistry and immunofluorescence and quantitative PCR. Results Synovial tissue of axSpA patients shows significantly more IL26-positive cells than that of HCs (p < 0.01), but numbers are also elevated in PsA and RA patients. Immunofluorescence shows co-localization of IL26 with CD68, but not with CD3, SMA, CD163, cadherin-11, or CD90. IL26 is elevated in the serum of RA and PsA (but not axSpA) patients compared with HCs (p < 0.001 and p < 0.01). However, peripheral blood CD4+ T cells from axSpA and PsA patients show higher positivity for IL26 in the PrimeFlow assay compared with HCs. CD4+ memory T cells from axSpA patients produce more IL26 under Th17-favoring conditions (IL-1β and IL-23) than cells from PsA and RA patients or HCs. Conclusion IL26 production is increased in the synovial tissue of SpA and can be localized to CD68+ macrophage-like synoviocytes, whereas circulating IL26+ Th17 cells are only modestly enriched. Considering the osteoproliferative properties of IL26, this offers new therapeutic options independent of Th17 pathways.
Collapse
Affiliation(s)
- Ariane Hammitzsch
- Department of Nephrology, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Andreas Ossadnik
- Department of Nephrology, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Quirin Bachmann
- Department of Nephrology, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Helga Merwald-Fraenk
- Amedes Holding AG, Ambulatory Healthcare Center (MVZ) Endokrinologikum München, Munich, Germany
| | - Georg Lorenz
- Department of Nephrology and Rheumatology, Klinik Augustinum München, Munich, Germany
| | | | - Franziska Wiesent
- Amedes Holding AG, Ambulatory Healthcare Center (MVZ) Endokrinologikum München, Munich, Germany
| | - Heinrich Mühlhofer
- Clinic and Policlinic of Orthopaedics and Sports’ Orthopaedics, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Davide Simone
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Paul Bowness
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Uwe Heemann
- Department of Nephrology, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Martin Arbogast
- Department of Rheumatic Orthopedics and Hand Surgery, Klinik Oberammergau, Waldburg-Zeil Kliniken GmbH und Co KG, Oberammergau, Germany
| | - Philipp Moog
- Department of Nephrology, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Christoph Schmaderer
- Department of Nephrology, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
4
|
Müller S, Kröger C, Schultze JL, Aschenbrenner AC. Whole blood stimulation as a tool for studying the human immune system. Eur J Immunol 2024; 54:e2350519. [PMID: 38103010 DOI: 10.1002/eji.202350519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 12/05/2023] [Accepted: 12/07/2023] [Indexed: 12/17/2023]
Abstract
The human immune system is best accessible via tissues and organs not requiring major surgical intervention, such as blood. In many circumstances, circulating immune cells correlate with an individual's health state and give insight into physiological and pathophysiological processes. Stimulating whole blood ex vivo is a powerful tool to investigate immune responses. In the context of clinical research, the applications of whole blood stimulation include host immunity, disease characterization, diagnosis, treatment, and drug development. Here, we summarize different setups and readouts of whole blood assays and discuss applications for preclinical research and clinical practice. Finally, we propose combining whole blood stimulation with high-throughput technologies, such as single-cell RNA-sequencing, to comprehensively analyze the human immune system for the identification of biomarkers, therapeutic interventions as well as companion diagnostics.
Collapse
Affiliation(s)
- Sophie Müller
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) e.V., Bonn, Germany
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Genomics & Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Charlotte Kröger
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) e.V., Bonn, Germany
- Genomics & Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Joachim L Schultze
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) e.V., Bonn, Germany
- Genomics & Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, DZNE and University of Bonn, Bonn, Germany
| | - Anna C Aschenbrenner
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) e.V., Bonn, Germany
| |
Collapse
|
5
|
Møller DL, Sørensen SS, Perch M, Gustafsson F, Hald A, Knudsen AD, Abdulovski R, Arentoft NS, Lundgren J, Rasmussen A, Ostrowski SR, Nielsen SD. Differences in toll-like receptor ligand-induced cytokine concentrations before and after solid organ transplantation: A prospective, observational cohort study in a clinical setting. Scand J Immunol 2024; 99:e13337. [PMID: 38168873 DOI: 10.1111/sji.13337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/09/2023] [Accepted: 10/20/2023] [Indexed: 01/05/2024]
Abstract
Reliable methods to assess immune function after solid organ transplantation (SOT) are needed to guide dosing of immunosuppression. We hypothesized that toll-like receptor ligand-induced cytokine concentrations would decrease post-transplantation due to the use of immunosuppressive medication. Furthermore, we hypothesized that induced cytokine concentrations pre-transplantation would be higher in recipients with episodes of acute rejection post-transplantation due to underlying immunological dispositions. We aimed to investigate toll-like receptor ligand-induced cytokine concentrations by TruCulture©, a standardized immunoassay, in SOT recipients before and 3 months after SOT and explored associations with methylprednisolone-treated acute rejections. We conducted a prospective, observational cohort study including 123 participants (67 liver, 32 kidney and 24 lung transplant recipients). Whole blood was stimulated for 22 h with: (A) Lipopolysaccharide (LPS), (B) Resiquimod, (C) Polyinosinic:polycytidylic acid (Poly I:C) and (D) a blank control. Cytokine concentrations (TNF-α, IL-1β, IL-6, IL-8, IL-10, IL-12p40, IL-17A, IFN-α and IFN-γ) were measured by Luminex. 30 participants developed methylprednisolone-treated acute rejection at a median of 9 days (IQR 5-17) post-SOT. We found that all induced cytokine concentrations decreased post-SOT except from LPS-induced and Poly I:C-induced IL-10. The induced cytokine concentration pre-transplantation did not differ in recipients with or without acute rejection. In conclusion, the induced cytokine concentrations decreased for all stimuli post-SOT, except the anti-inflammatory cytokine IL-10. Importantly, recipients developing early acute rejection did not differ in induced cytokine concentrations pre-SOT. Thus, the use of a standardized assay in SOT is feasible in a clinical setting and may provide important information on the immune function post-SOT.
Collapse
Affiliation(s)
- Dina Leth Møller
- Viro-immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Søren Schwartz Sørensen
- Department of Nephrology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Michael Perch
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Cardiology, Section for Lung Transplantation, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Finn Gustafsson
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Annemette Hald
- Viro-immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Andreas Delhbaek Knudsen
- Viro-immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ranya Abdulovski
- Viro-immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Nicoline Stender Arentoft
- Viro-immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jens Lundgren
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Centre of Excellence for Health, Immunity, and Infections, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Allan Rasmussen
- Department of Surgical Gastroenterology and Transplantation, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Sisse Rye Ostrowski
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Immunology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Susanne Dam Nielsen
- Viro-immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Surgical Gastroenterology and Transplantation, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
6
|
Yeh YT, Tsai TF. Drug- or Vaccine-Induced/Aggravated Psoriatic Arthritis: A Systematic Review. Dermatol Ther (Heidelb) 2024; 14:59-81. [PMID: 38183617 PMCID: PMC10828154 DOI: 10.1007/s13555-023-01082-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 12/05/2023] [Indexed: 01/08/2024] Open
Abstract
INTRODUCTION Drugs and vaccines have been less studied as inducing or aggravating factors for psoriatic arthritis (PsA) compared with psoriasis. Thus, the present study collected and summarized the publications to date about this issue. METHODS We conducted a systematic literature search through the PubMed, Embase, and Cochrane databases to identify all reports on potential drug- and vaccine-related PsA events until 28 February 2023. RESULTS In total, 179 cases from 79 studies were eligible for study. Drugs commonly reported include coronavirus disease 2019 (COVID-19) mRNA vaccines (6 cases), bacillus Calmette-Guerin (BCG) vaccine (3 cases), interferon (18 cases), immune-checkpoint inhibitors (ICI) (19 cases), and biologic disease-modifying antirheumatic drugs (bDMARDs) (127 cases). Drugs causing psoriasis may also induce or aggravate PsA (6 cases). BDMARD-related PsA mostly occurred in a "paradoxical" setting, in which the bDMARDs approved for the treatment of psoriasis induce or aggravate PsA. The reported latency may be delayed up to 2 years. Peripheral arthritis (82.3%) was the most common manifestation of drug- and vaccine-related PsA, followed by dactylitis (29.1%), enthesitis (23.4%), and spondyloarthritis (17.7%). CONCLUSIONS Drugs and vaccines may be implicated in the aggravation of PsA. Possible mechanisms include cytokine imbalance, immune dysregulation, or inadequate PsA treatment response compared with psoriasis. Most reports are case based without controls, so more studies are needed to further prove the causality. However, early recognition of factors causing or aggravating PsA is important to prevent the irreversible joint damage.
Collapse
Affiliation(s)
- Yao-Tsung Yeh
- Department of Medical Education, National Taiwan University Hospital, Taipei, Taiwan
| | - Tsen-Fang Tsai
- Department of Dermatology, National Taiwan University Hospital, No. 7, Chung Shan South Road (Zhongshan S. Rd.), Zhongzheng Dist., Taipei City, 100225, Taiwan, ROC.
| |
Collapse
|
7
|
Kim SH, Lee SH. Updates on ankylosing spondylitis: pathogenesis and therapeutic agents. JOURNAL OF RHEUMATIC DISEASES 2023; 30:220-233. [PMID: 37736590 PMCID: PMC10509639 DOI: 10.4078/jrd.2023.0041] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 09/23/2023]
Abstract
Ankylosing spondylitis (AS) is an autoinflammatory disease that manifests with the unique feature of enthesitis. Gut microbiota, HLA-B*27, and biomechanical stress mutually influence and interact resulting in setting off a flame of inflammation. In the HLA-B*27 positive group, dysbiosis in the gut environment disrupts the barrier to exogenous bacteria or viruses. Additionally, biomechanical stress induces inflammation through enthesial resident or gut-origin immune cells. On this basis, innate and adaptive immunity can propagate inflammation and lead to chronic disease. Finally, bone homeostasis is regulated by cytokines, by which the inflamed region is substituted into new bone. Agents that block cytokines are constantly being developed to provide diverse therapeutic options for preventing the progression of inflammation. In addition, some antibodies have been shown to distinguish disease selectively, which support the involvement of autoimmune immunity in AS. In this review, we critically analyze the complexity and uniqueness of the pathogenesis with updates on the findings of immunity and provide new information about biologics and biomarkers.
Collapse
Affiliation(s)
- Se Hee Kim
- Division of Rheumatology, Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Sang-Hoon Lee
- Division of Rheumatology, Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, College of Medicine, Kyung Hee University, Seoul, Korea
| |
Collapse
|
8
|
Lee BW, Moon SJ. Inflammatory Cytokines in Psoriatic Arthritis: Understanding Pathogenesis and Implications for Treatment. Int J Mol Sci 2023; 24:11662. [PMID: 37511421 PMCID: PMC10381020 DOI: 10.3390/ijms241411662] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/16/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Psoriatic arthritis (PsA) is a persistent, inflammatory disease that affects individuals with psoriasis, arthritis, and enthesitis. Research has demonstrated that inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α), interleukin-23 (IL-23), and interleukin-17 (IL-17) play a pivotal role in both the onset and progression of PsA. These cytokines are generated by activated immune cells and stimulate the attraction of inflammatory cells to the synovium and joint tissues, resulting in the deterioration of cartilage and bone. The blocking of these cytokines has become a successful treatment strategy for PsA, as biological drugs that inhibit TNF-α, IL-23, and IL-17 have demonstrated notable clinical benefits. The association between PsA and other types of inflammatory cytokines or chemokines, excluding TNF-α, IL-23, and IL-17, has been extensively investigated in numerous studies. These findings may provide a chance for the discovery of novel therapeutic agents targeting other molecules, distinct from the currently approved biologics and targeted synthetic disease-modifying anti-rheumatic drugs. In this review, we discuss the current understanding of the role of inflammatory cytokines in PsA pathogenesis and clinical implications of targeting these cytokines for PsA treatment.
Collapse
Affiliation(s)
- Bong-Woo Lee
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Su-Jin Moon
- Division of Rheumatology, Department of Internal Medicine, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 07345, Republic of Korea
| |
Collapse
|
9
|
Chen J, Zhong H, Yu H, Sun J, Shen B, Xu X, Huang S, Huang P, Zhong Y. Interleukin-17A modulates retinal inflammation by regulating microglial activation via the p38 MAPK pathway in experimental glaucoma neuropathy. FASEB J 2023; 37:e22945. [PMID: 37144630 DOI: 10.1096/fj.202202056rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 04/10/2023] [Accepted: 04/18/2023] [Indexed: 05/06/2023]
Abstract
As a prototypical member of the IL-17 family, interleukin-17A (IL-17A) has received increasing attentions for its potent proinflammatory role as well as potential to be a key therapeutic target in human autoimmune inflammatory diseases; however, its roles in other pathological scenarios like neuroinflammations are not fully elucidated yet but appear essentially correlating and promising. Glaucoma is the leading cause of irreversible blindness with complicated pathogenesis still to be understood, where neuroinflammation was reported to be critically involved in its both initiation and progression. Whether IL-17A takes part in the pathogenesis of glaucoma through interfering neuroinflammation due to its potent proinflammatory effect is still unknown. In the present study, we investigated the role of IL-17A in the pathological process of glaucoma neuropathy as well as its relationship with the predominant immune inflammation mediator microglia in retina, trying to elucidate the underlying mechanisms from the view of inflammation modulation. In our study, RNA sequencing was performed for the retinas of chronic ocular hypertension (COH) and control mice. Western blot, RT-PCR, immunofluorescence, and ELISA were used to evaluate the microglial activation and proinflammatory cytokines release at conditioned levels of IL-17A, along with assessment of optic nerve integrity including retinal ganglion cells (RGCs) counting, axonal neurofilament quantification, and flash visual-evoked potential (F-VEP) examination. And the possibly involved signaling pathways were screened out to go through further validation in scenarios with conditioned IL-17A. Subsequently, IL-17A was found to be significantly upregulated in COH retina. Furthermore, suppression of IL-17A effectively diminished the loss of RGCs, improved axonal quality, and F-VEP performance in COH mice. Mechanistically, IL-17A promoted microglial activation and proinflammatory cytokines release along with enhanced phenotypic conversion of activated microglia to M2-type in early stage and to M1-type in late stage in glaucomatous retinas. Microglia elimination decreased the proinflammatory factors secretion, enhanced the RGCs survival and axonal quality mediated by IL-17A. Furthermore, IL-17A-induced the overactivation of microglia in glaucomatous condition was alleviated after blocking the p38 MAPK pathway. Taken together, IL-17A is involved in the regulation of retinal immune response and RGCs cell death in experimental glaucoma by essentially promoting retinal microglial activation via p38 MAPK signaling pathway. IL-17A dynamically regulates the phenotypic conversion of retinal microglia in experimental glaucoma partly depending on the duration of elevated intraocular pressure. Suppression of IL-17A contributes to alleviate glaucoma neuropathy and exhibits promising potential as an innovative target for therapeutic strategy in glaucoma.
Collapse
Affiliation(s)
- Junjue Chen
- Department of Ophthalmology, Ruijin Hospital Affiliated Medical School, Shanghai Jiao Tong University, Shanghai, China
| | - Huimin Zhong
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huan Yu
- Department of Ophthalmology, Ruijin Hospital Affiliated Medical School, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Sun
- Department of Ophthalmology, Ruijin Hospital Affiliated Medical School, Shanghai Jiao Tong University, Shanghai, China
| | - Bingqiao Shen
- Department of Ophthalmology, Ruijin Hospital Affiliated Medical School, Shanghai Jiao Tong University, Shanghai, China
| | - Xing Xu
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital Affiliated Medical School, Shanghai Jiao Tong University, Shanghai, China
| | - Shouyue Huang
- Department of Ophthalmology, Ruijin Hospital Affiliated Medical School, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Huang
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital Affiliated Medical School, Shanghai Jiao Tong University, Shanghai, China
| | - Yisheng Zhong
- Department of Ophthalmology, Ruijin Hospital Affiliated Medical School, Shanghai Jiao Tong University, Shanghai, China
- Department of Ophthalmology, Zhoushan Branch of Ruijin Hospital Affiliated Medical School, Shanghai Jiao Tong University, Zhoushan, China
| |
Collapse
|
10
|
Ciliento MS, Venturelli V, Schettini N, Bertola R, Garaffoni C, Lanza G, Gafà R, Borghi A, Corazza M, Zabotti A, Missiroli S, Boncompagni C, Patergnani S, Perrone M, Giorgi C, Pinton P, Govoni M, Scirè CA, Bortoluzzi A, Silvagni E. Evaluation of the Synovial Effects of Biological and Targeted Synthetic DMARDs in Patients with Psoriatic Arthritis: A Systematic Literature Review and Meta-Analysis. Int J Mol Sci 2023; 24:5006. [PMID: 36902437 PMCID: PMC10002880 DOI: 10.3390/ijms24055006] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/26/2023] [Accepted: 02/27/2023] [Indexed: 03/08/2023] Open
Abstract
The aims of this systematic literature review (SLR) were to identify the effects of approved biological and targeted synthetic disease modifying antirheumatic drugs (b/tsDMARDs) on synovial membrane of psoriatic arthritis (PsA) patients, and to determine the existence of histological/molecular biomarkers of response to therapy. A search was conducted on MEDLINE, Embase, Scopus, and Cochrane Library (PROSPERO:CRD42022304986) to retrieve data on longitudinal change of biomarkers in paired synovial biopsies and in vitro studies. A meta-analysis was conducted by adopting the standardized mean difference (SMD) as a measure of the effect. Twenty-two studies were included (19 longitudinal, 3 in vitro). In longitudinal studies, TNF inhibitors were the most used drugs, while, for in vitro studies, JAK inhibitors or adalimumab/secukinumab were assessed. The main technique used was immunohistochemistry (longitudinal studies). The meta-analysis showed a significant reduction in both CD3+ lymphocytes (SMD -0.85 [95% CI -1.23; -0.47]) and CD68+ macrophages (sublining, sl) (SMD -0.74 [-1.16; -0.32]) in synovial biopsies from patients treated for 4-12 weeks with bDMARDs. Reduction in CD3+ mostly correlated with clinical response. Despite heterogeneity among the biomarkers evaluated, the reduction in CD3+/CD68+sl cells during the first 3 months of treatment with TNF inhibitors represents the most consistent variation reported in the literature.
Collapse
Affiliation(s)
- Maria Sofia Ciliento
- Rheumatology Unit, Department of Medical Sciences, Università degli Studi di Ferrara, Azienda Ospedaliero-Universitaria S. Anna, 44124 Cona, Italy
- Department of Precision Medicine, University della Campania L. Vanvitelli, 80138 Naples, Italy
| | - Veronica Venturelli
- Rheumatology Unit, Department of Medical Sciences, Università degli Studi di Ferrara, Azienda Ospedaliero-Universitaria S. Anna, 44124 Cona, Italy
| | - Natale Schettini
- Section of Dermatology, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Riccardo Bertola
- Rheumatology Unit, Department of Medical Sciences, Università degli Studi di Ferrara, Azienda Ospedaliero-Universitaria S. Anna, 44124 Cona, Italy
| | - Carlo Garaffoni
- Rheumatology Unit, Department of Medical Sciences, Università degli Studi di Ferrara, Azienda Ospedaliero-Universitaria S. Anna, 44124 Cona, Italy
| | - Giovanni Lanza
- Anatomic Pathology, Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Roberta Gafà
- Anatomic Pathology, Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Alessandro Borghi
- Section of Dermatology, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Monica Corazza
- Section of Dermatology, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Alen Zabotti
- Department of Medical and Biological Sciences, Rheumatology Institute, University Hospital Santa Maria della Misericordia, 33100 Udine, Italy
| | - Sonia Missiroli
- Laboratory for Technologies of Advanced Therapies, Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Caterina Boncompagni
- Laboratory for Technologies of Advanced Therapies, Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Simone Patergnani
- Laboratory for Technologies of Advanced Therapies, Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Mariasole Perrone
- Laboratory for Technologies of Advanced Therapies, Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Carlotta Giorgi
- Laboratory for Technologies of Advanced Therapies, Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Paolo Pinton
- Laboratory for Technologies of Advanced Therapies, Department of Medical Sciences, Section of Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Marcello Govoni
- Rheumatology Unit, Department of Medical Sciences, Università degli Studi di Ferrara, Azienda Ospedaliero-Universitaria S. Anna, 44124 Cona, Italy
| | - Carlo Alberto Scirè
- School of Medicine, University of Milano Bicocca, 20126 Milan, Italy
- Epidemiology Unit, Italian Society for Rheumatology, 20121 Milan, Italy
| | - Alessandra Bortoluzzi
- Rheumatology Unit, Department of Medical Sciences, Università degli Studi di Ferrara, Azienda Ospedaliero-Universitaria S. Anna, 44124 Cona, Italy
| | - Ettore Silvagni
- Rheumatology Unit, Department of Medical Sciences, Università degli Studi di Ferrara, Azienda Ospedaliero-Universitaria S. Anna, 44124 Cona, Italy
| |
Collapse
|
11
|
Phenotypic heterogeneity in psoriatic arthritis: towards tissue pathology-based therapy. Nat Rev Rheumatol 2023; 19:153-165. [PMID: 36596924 DOI: 10.1038/s41584-022-00874-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2022] [Indexed: 01/04/2023]
Abstract
Psoriatic arthritis (PsA) is a heterogeneous disease involving multiple potential tissue domains. Most outcome measures used so far in randomized clinical trials do not sufficiently reflect this domain heterogeneity. The concept that pathogenetic mechanisms might vary across tissues within a single disease, underpinning such phenotype diversity, could explain tissue-distinct levels of response to different therapies. In this Review, we discuss the tissue, cellular and molecular mechanisms that drive clinical heterogeneity in PsA phenotypes, and detail existing tissue-based research, including data generated using sophisticated interrogative technologies with single-cell precision. Finally, we discuss how these elements support the need for tissue-based therapy in PsA in the context of existing and new therapeutic modes of action, and the implications for future PsA trial outcomes and design.
Collapse
|
12
|
Paroli M, Spadea L, Caccavale R, Spadea L, Paroli MP, Nante N. The Role of Interleukin-17 in Juvenile Idiopathic Arthritis: From Pathogenesis to Treatment. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:1552. [PMID: 36363508 PMCID: PMC9696590 DOI: 10.3390/medicina58111552] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/22/2022] [Accepted: 10/26/2022] [Indexed: 04/12/2024]
Abstract
Background and Objectives: Interleukin-17 (IL-17) is a cytokine family consisting of six members and five specific receptors. IL-17A was the first member to be identified in 1993. Since then, several studies have elucidated that IL-17 has predominantly pro-inflammatory activity and that its production is involved in both the defense against pathogens and the genesis of autoimmune processes. Materials and Methods: In this review, we provide an overview of the role of interleukin-17 in the pathogenesis of juvenile idiopathic arthritis (JIA) and its relationship with IL-23, the so-called IL-23-IL-17 axis, by reporting updated findings from the scientific literature. Results: Strong evidence supports the role of interleukin-17A in the pathogenesis of JIA after the deregulated production of this interleukin by both T helper 17 (Th17) cells and cells of innate immunity. The blocking of IL-17A was found to improve the course of JIA, leading to the approval of the use of the human anti-IL17A monoclonal antibody secukinumab in the treatment of the JIA subtypes juvenile psoriatic arthritis (JPsA) and enthesitis-related arthritis (ERA). Conclusions: IL-17A plays a central role in the pathogenesis of JIA. Blocking its production with specific biologic drugs enables the effective treatment of this disabling childhood rheumatic disease.
Collapse
Affiliation(s)
- Marino Paroli
- Division of Clinical Immunology, Department of Clinical, Anesthesiologic and Cardiovascular Sciences, Faculty of Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Luca Spadea
- Post Graduate School of Public Health, University of Siena, 53100 Siena, Italy
| | - Rosalba Caccavale
- Division of Clinical Immunology, Department of Clinical, Anesthesiologic and Cardiovascular Sciences, Faculty of Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Leopoldo Spadea
- Eye Clinic, Department of Sense Organs, Sapienza University of Rome, 00185 Rome, Italy
| | - Maria Pia Paroli
- Eye Clinic, Department of Sense Organs, Sapienza University of Rome, 00185 Rome, Italy
| | - Nicola Nante
- Post Graduate School of Public Health, University of Siena, 53100 Siena, Italy
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy
| |
Collapse
|
13
|
Martínez-Ramos S, Rafael-Vidal C, Pego-Reigosa JM, García S. Monocytes and Macrophages in Spondyloarthritis: Functional Roles and Effects of Current Therapies. Cells 2022; 11:cells11030515. [PMID: 35159323 PMCID: PMC8834543 DOI: 10.3390/cells11030515] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 01/27/2023] Open
Abstract
Spondyloarthritis (SpA) is a family of chronic inflammatory diseases, being the most prevalent ankylosing spondylitis (AS) and psoriatic arthritis (PsA). These diseases share genetic, clinical and immunological features, such as the implication of human leukocyte antigen (HLA) class I molecule 27 (HLA-B27), the inflammation of peripheral, spine and sacroiliac joints and the presence of extra-articular manifestations (psoriasis, anterior uveitis, enthesitis and inflammatory bowel disease). Monocytes and macrophages are essential cells of the innate immune system and are the first line of defence against external agents. In rheumatic diseases including SpA, the frequency and phenotypic and functional characteristics of both cell types are deregulated and are involved in the pathogenesis of these diseases. In fact, monocytes and macrophages play key roles in the inflammatory processes characteristics of SpA. The aim of this review is analysing the characteristics and functional roles of monocytes and macrophages in these diseases, as well as the impact of different current therapies on these cell types.
Collapse
Affiliation(s)
- Sara Martínez-Ramos
- Rheumatology & Immuno-Mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, 36312 Vigo, Spain; (S.M.-R.); (C.R.-V.); (J.M.P.-R.)
- Rheumatology Department, University Hospital Complex of Vigo, 36214 Vigo, Spain
| | - Carlos Rafael-Vidal
- Rheumatology & Immuno-Mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, 36312 Vigo, Spain; (S.M.-R.); (C.R.-V.); (J.M.P.-R.)
- Rheumatology Department, University Hospital Complex of Vigo, 36214 Vigo, Spain
| | - José M. Pego-Reigosa
- Rheumatology & Immuno-Mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, 36312 Vigo, Spain; (S.M.-R.); (C.R.-V.); (J.M.P.-R.)
- Rheumatology Department, University Hospital Complex of Vigo, 36214 Vigo, Spain
| | - Samuel García
- Rheumatology & Immuno-Mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, 36312 Vigo, Spain; (S.M.-R.); (C.R.-V.); (J.M.P.-R.)
- Rheumatology Department, University Hospital Complex of Vigo, 36214 Vigo, Spain
- Correspondence: ; Tel.: +34-986-217-463
| |
Collapse
|
14
|
Bolt JW, van Kuijk AW, Teunissen MBM, van der Coelen D, Aarrass S, Gerlag DM, Tak PP, van de Sande MG, Lebre MC, van Baarsen LGM. Impact of Adalimumab Treatment on Interleukin-17 and Interleukin-17 Receptor Expression in Skin and Synovium of Psoriatic Arthritis Patients with Mild Psoriasis. Biomedicines 2022; 10:biomedicines10020324. [PMID: 35203534 PMCID: PMC8869729 DOI: 10.3390/biomedicines10020324] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/19/2022] [Accepted: 01/24/2022] [Indexed: 02/04/2023] Open
Abstract
Interleukin (IL)-17 and tumor necrosis factor-alpha (TNF)-α are key players in psoriatic arthritis (PsA) pathogenesis. While both cytokines can be therapeutically targeted with beneficial clinical outcome, it is unclear whether inhibiting one cytokine will affect the other at sites of inflammation. If both act independently, this might provide a rationale for dual or combined inhibition of both cytokines. Here, we evaluated the effect of TNF blockade in PsA patients on IL-17 levels in both skin and synovial tissue biopsies. PsA patients with mild psoriatic skin lesions were randomized to receive either adalimumab or placebo for four weeks. Synovial and skin biopsies were obtained at weeks zero and four. Skin from healthy donors (HDs) was used for comparison. Expression of IL-17A, IL-17F, IL-17RA and IL-17RC was assessed by immunohistochemistry and analyzed with digital image analysis. We found relatively low levels of IL-17 and its receptors in the skin of PsA patients compared to HD, and only IL-17F in the dermis of lesional psoriatic skin was significantly higher compared to HD skin (p = 0.0002). Histologically IL-17A, IL-17F, IL-17RA and IL-17RC in skin and synovial tissue were not downregulated by adalimumab treatment. Thus, in this cohort of PsA patients with mild psoriasis, TNF blockade did not affect the protein levels of IL-17 cytokines and its receptors in skin and synovium, despite reduced cellular inflammation and improved clinical outcome for joint involvement.
Collapse
Affiliation(s)
- Janne W. Bolt
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (J.W.B.); (D.v.d.C.); (S.A.); (D.M.G.); (P.P.T.); (M.G.v.d.S.)
- Department of Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC)-Reade, 1040 HG Amsterdam, The Netherlands;
| | - Arno W. van Kuijk
- Department of Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC)-Reade, 1040 HG Amsterdam, The Netherlands;
- Department of Rheumatology, Reade, 1056 AB Amsterdam, The Netherlands
| | - Marcel B. M. Teunissen
- Department of Dermatology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Dennis van der Coelen
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (J.W.B.); (D.v.d.C.); (S.A.); (D.M.G.); (P.P.T.); (M.G.v.d.S.)
- Department of Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC)-Reade, 1040 HG Amsterdam, The Netherlands;
| | - Saïda Aarrass
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (J.W.B.); (D.v.d.C.); (S.A.); (D.M.G.); (P.P.T.); (M.G.v.d.S.)
- Department of Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC)-Reade, 1040 HG Amsterdam, The Netherlands;
| | - Daniëlle M. Gerlag
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (J.W.B.); (D.v.d.C.); (S.A.); (D.M.G.); (P.P.T.); (M.G.v.d.S.)
- Department of Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC)-Reade, 1040 HG Amsterdam, The Netherlands;
| | - Paul P. Tak
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (J.W.B.); (D.v.d.C.); (S.A.); (D.M.G.); (P.P.T.); (M.G.v.d.S.)
- Department of Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC)-Reade, 1040 HG Amsterdam, The Netherlands;
- Candel Therapeutics, Needham, MA 02494, USA
- Internal Medicine, Cambridge University, Cambridge CB2 1TN, UK
| | - Marleen G. van de Sande
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (J.W.B.); (D.v.d.C.); (S.A.); (D.M.G.); (P.P.T.); (M.G.v.d.S.)
- Department of Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC)-Reade, 1040 HG Amsterdam, The Netherlands;
| | - Maria C. Lebre
- The Netherlands Cancer Institute, Division of Pharmacology, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands;
| | - Lisa G. M. van Baarsen
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (J.W.B.); (D.v.d.C.); (S.A.); (D.M.G.); (P.P.T.); (M.G.v.d.S.)
- Department of Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC)-Reade, 1040 HG Amsterdam, The Netherlands;
- Correspondence:
| |
Collapse
|
15
|
Kang S, Wu Q, Yang B, Wu C. Estrogen enhanced the expression of IL‐17 by tissue‐resident memory γδT cells from uterus via interferon regulatory factor 4. FASEB J 2022; 36:e22166. [DOI: 10.1096/fj.202101443rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/14/2021] [Accepted: 12/29/2021] [Indexed: 11/11/2022]
Affiliation(s)
- Shuangpeng Kang
- Academician Workstation Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations Changsha Medical University Changsha P.R. China
| | - Qiongli Wu
- Institute of Immunology Zhongshan School of Medicine Sun Yat‐sen University Guangzhou P.R. China
| | - Binyan Yang
- Institute of Immunology Zhongshan School of Medicine Sun Yat‐sen University Guangzhou P.R. China
| | - Changyou Wu
- Institute of Immunology Zhongshan School of Medicine Sun Yat‐sen University Guangzhou P.R. China
| |
Collapse
|
16
|
Hammoura I, Fiechter RH, Bryant SH, Westmoreland S, Kingsbury G, Waegell W, Tas SW, Baeten DL, van de Sande MGH, van Tok MN, van Duivenvoorde LM. Dual Blockade of TNF and IL-17A Inhibits Inflammation and Structural Damage in a Rat Model of Spondyloarthritis. Int J Mol Sci 2022; 23:ijms23020859. [PMID: 35055042 PMCID: PMC8776047 DOI: 10.3390/ijms23020859] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/07/2022] [Accepted: 01/08/2022] [Indexed: 12/20/2022] Open
Abstract
The tumor necrosis factor (TNF) and IL-23/IL-17 axes are the main therapeutic targets in spondyloarthritis. Despite the clinical efficacy of blocking either pathway, monotherapy does not induce remission in all patients and its effect on new bone formation remains unclear. We aimed to study the effect of TNF and IL-17A dual inhibition on clinical disease and structural damage using the HLA-B27/human β2-microglobulin transgenic rat model of SpA. Immunized rats were randomized according to arthritis severity, 1 week after arthritis incidence reached 50%, to be treated twice weekly for a period of 5 weeks with either a dual blockade therapy of an anti-TNF antibody and an anti-IL-17A antibody, a single therapy of either antibody, or PBS as vehicle control. Treatment-blinded observers assessed inflammation and structural damage clinically, histologically and by micro-CT imaging. Both single therapies as well as TNF and IL-17A dual blockade therapy reduced clinical spondylitis and peripheral arthritis effectively and similarly. Clinical improvement was confirmed for all treatments by a reduction of histological inflammation and pannus formation (p < 0.05) at the caudal spine. All treatments showed an improvement of structural changes at the axial and peripheral joints on micro-CT imaging, with a significant decrease for roughness (p < 0.05), which reflects both erosion and new bone formation, at the level of the caudal spine. The effect of dual blockade therapy on new bone formation was more prominent at the axial than the peripheral level. Collectively, our study showed that dual blockade therapy significantly reduces inflammation and structural changes, including new bone formation. However, we could not confirm a more pronounced effect of dual inhibition compared to single inhibition.
Collapse
Affiliation(s)
- Ihsan Hammoura
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology and Rheumatology, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (I.H.); (R.H.F.); (S.W.T.); (D.L.B.); (M.N.v.T.); (L.M.v.D.)
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Renee H. Fiechter
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology and Rheumatology, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (I.H.); (R.H.F.); (S.W.T.); (D.L.B.); (M.N.v.T.); (L.M.v.D.)
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Shaughn H. Bryant
- AbbVie Bioresearch Center, Worcester, MA 01605, USA; (S.H.B.); (S.W.); (G.K.); (W.W.)
| | - Susan Westmoreland
- AbbVie Bioresearch Center, Worcester, MA 01605, USA; (S.H.B.); (S.W.); (G.K.); (W.W.)
| | - Gillian Kingsbury
- AbbVie Bioresearch Center, Worcester, MA 01605, USA; (S.H.B.); (S.W.); (G.K.); (W.W.)
| | - Wendy Waegell
- AbbVie Bioresearch Center, Worcester, MA 01605, USA; (S.H.B.); (S.W.); (G.K.); (W.W.)
| | - Sander W. Tas
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology and Rheumatology, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (I.H.); (R.H.F.); (S.W.T.); (D.L.B.); (M.N.v.T.); (L.M.v.D.)
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Dominique L. Baeten
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology and Rheumatology, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (I.H.); (R.H.F.); (S.W.T.); (D.L.B.); (M.N.v.T.); (L.M.v.D.)
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Marleen G. H. van de Sande
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology and Rheumatology, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (I.H.); (R.H.F.); (S.W.T.); (D.L.B.); (M.N.v.T.); (L.M.v.D.)
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Correspondence:
| | - Melissa N. van Tok
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology and Rheumatology, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (I.H.); (R.H.F.); (S.W.T.); (D.L.B.); (M.N.v.T.); (L.M.v.D.)
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Leonie M. van Duivenvoorde
- Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology and Rheumatology, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (I.H.); (R.H.F.); (S.W.T.); (D.L.B.); (M.N.v.T.); (L.M.v.D.)
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
17
|
Elliott A, McGonagle D, Rooney M. Integrating imaging and biomarker assessment to better define psoriatic arthritis and predict response to biologic therapy. Rheumatology (Oxford) 2021; 60:vi38-vi52. [PMID: 34951926 PMCID: PMC8709569 DOI: 10.1093/rheumatology/keab504] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 06/08/2021] [Indexed: 12/13/2022] Open
Abstract
The treatment options for PsA have substantially expanded over the last decade. Approximately 40% of patients will not respond to first-line anti-TNF-α therapies. There is limited data to help clinicians select the most appropriate biologic therapy for PsA patients, including guidance for decisions on biologic therapy switching. In this review we will examine the current understanding of predictors of response to treatment. Imaging technology has evolved to allow us to better study psoriatic disease and define disease activity, including synovitis and enthesitis. Enthesitis is implicated in the pathogenesis, diagnosis and prognosis of PsA. It appears to be a common thread among all of the various PsA clinical presentations. Enthesitis mainly manifests as tenderness, which is difficult to distinguish from FM, chronic pain and mechanically associated enthesopathy, and it might be relevant for understanding the apparent 40% failure of existing therapy. Excess adipose tissue makes if more difficult to detect joint swelling clinically, as many PsA patients have very high BMIs. Integrating imaging and clinical assessment with biomarker analysis could help to deliver stratified medicine in PsA and allow better treatment decision making. This could include which patients require ongoing biologic therapy, which class of biologic therapy that should be, and who alternatively requires management of non-inflammatory disease.
Collapse
Affiliation(s)
- Ashley Elliott
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast, UK
| | - Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- Leeds Musculoskeletal Biomedical Research Unit, Chapel Allerton Hospital, Leeds, UK
| | - Madeleine Rooney
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast, UK
| |
Collapse
|
18
|
Todberg T, Loft N, Møller DL, Ostrowski SR, Nielsen SD, Skov L. Impact of methotrexate and adalimumab on immune function of patients with psoriasis. Dermatol Ther 2021; 35:e15284. [PMID: 34953013 DOI: 10.1111/dth.15284] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/01/2021] [Accepted: 12/16/2021] [Indexed: 11/26/2022]
Abstract
Patients with psoriasis are commonly treated with methotrexate or biologics. We examined the immune response in a whole blood assay (TruCulture®) to assess the effect of methotrexate and adalimumab. Twenty patients with psoriasis were included and cytokine levels following stimulation with LPS, R848, HKCA, PolyIC, or a blank were investigated before and after 3-6 months of treatment with methotrexate or adalimumab and in patients who had received adalimumab > 5 years. Methotrexate only induced minor changes in the cytokine responses, whereas adalimumab affected a wide range of cytokines important for the immune defense towards microorganisms. In the long-term adalimumab treated group, the cytokine levels were almost equivalent to the short-term adalimumab-treated group. Overall, methotrexate was not associated with cytokine suppression. Short and long-term treatment with adalimumab lowered multiple cytokines involved in the immune defense equally emphasizing the need to continuously be aware of the risk of infections in these patients. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Tanja Todberg
- Department of Dermatology and Allergy, Herlev and Gentofte Hospital, University of Copenhagen, DK-2900, Hellerup, Denmark.,Copenhagen Research Group for Inflammatory Skin (CORGIS), Hellerup, Denmark
| | - Nikolai Loft
- Department of Dermatology and Allergy, Herlev and Gentofte Hospital, University of Copenhagen, DK-2900, Hellerup, Denmark.,Copenhagen Research Group for Inflammatory Skin (CORGIS), Hellerup, Denmark
| | - Dina Leth Møller
- Viro-immonology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, Copenhagen University Hospital
| | - Sisse Rye Ostrowski
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen
| | - Susanne Dam Nielsen
- Viro-immonology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, Copenhagen University Hospital.,Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet
| | - Lone Skov
- Department of Dermatology and Allergy, Herlev and Gentofte Hospital, University of Copenhagen, DK-2900, Hellerup, Denmark.,Copenhagen Research Group for Inflammatory Skin (CORGIS), Hellerup, Denmark.,Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet
| |
Collapse
|
19
|
Padhan P, Maikap D. Secukinumab therapy in reactive arthritis: Report of two cases. Mod Rheumatol Case Rep 2021; 6:22-24. [PMID: 34508266 DOI: 10.1093/mrcr/rxab014] [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: 05/19/2021] [Revised: 06/14/2021] [Accepted: 07/01/2021] [Indexed: 01/20/2023]
Abstract
Reactive arthritis (ReA) is an immune-mediated aseptic synovitis resulting either from genitourinary or gastrointestinal tract, commonly presenting as oligoarthritis of the lower limbs and rarely urethritis and conjunctivitis. The treatment options include nonsteroidal anti-inflammatory drugs, conventional disease-modifying antirheumatic drugs, and biologics in severe cases. We report successful use of secukinumab in two cases of chronic severe ReA who initially failed to treatment with tumour necrosis factor inhibitor.
Collapse
Affiliation(s)
- Prasanta Padhan
- Department of Clinical Immunology and Rheumatology, Kalinga Institute of Medical Sciences, KIIT University, Bhubaneswar, Odisha, India
| | - Debashis Maikap
- Department of Clinical Immunology and Rheumatology, Kalinga Institute of Medical Sciences, KIIT University, Bhubaneswar, Odisha, India
| |
Collapse
|
20
|
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
|
21
|
Silvagni E, Missiroli S, Perrone M, Patergnani S, Boncompagni C, Bortoluzzi A, Govoni M, Giorgi C, Alivernini S, Pinton P, Scirè CA. From Bed to Bench and Back: TNF-α, IL-23/IL-17A, and JAK-Dependent Inflammation in the Pathogenesis of Psoriatic Synovitis. Front Pharmacol 2021; 12:672515. [PMID: 34211394 PMCID: PMC8241099 DOI: 10.3389/fphar.2021.672515] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/18/2021] [Indexed: 12/18/2022] Open
Abstract
Psoriatic arthritis (PsA) is a chronic inflammatory immune-mediated disease with a burdensome impact on quality of life and substantial healthcare costs. To date, pharmacological interventions with different mechanisms of action, including conventional synthetic (cs), biological (b), and targeted synthetic (ts) disease-modifying antirheumatic drugs (DMARDs), have been proven efficacious, despite a relevant proportion of failures. The current approach in clinical practice and research is typically "predictive": the expected response is based on stratification according to clinical, imaging, and laboratory data, with a "heuristic" approach based on "trial and error". Several available therapeutic options target the TNF-α pathway, while others are directed against the IL-23/IL-17A axis. Janus kinase inhibitors (JAKis), instead, simultaneously block different pathways, endowing these drugs with a potentially "broad-spectrum" mechanism of action. It is not clear, however, whether targeting a specific pathway (e.g., TNF-α or the IL-23/IL-17 axis) could result in discordant effects over other approaches. In particular, in the case of "refractory to a treatment" patients, other pathways might be hyperactivated, with opposing, synergistic, or redundant biological significance. On the contrary, refractory states could be purely resistant to treatment as a whole. Since chronic synovitis is one of the primary targets of inflammation in PsA, synovial biomarkers could be useful in depicting specific biological characteristics of the inflammatory burden at the single-patient level, and despite not yet being implemented in clinical practice, these biomarkers might help in selecting the proper treatment. In this narrative review, we will provide an up-to-date overview of the knowledge in the field of psoriatic synovitis regarding studies investigating the relationships among different activated proinflammatory processes suitable for targeting by different available drugs. The final objective is to clarify the state of the art in the field of personalized medicine for psoriatic disease, aiming at moving beyond the current treatment schedules toward a patient-centered approach.
Collapse
Affiliation(s)
- Ettore Silvagni
- Rheumatology Unit, Department of Medical Sciences, Università degli Studi di Ferrara and Azienda Ospedaliero-Universitaria S. Anna, Cona, Italy
| | - Sonia Missiroli
- Department of Medical Sciences, Section of Experimental Medicine, Laboratory for Technologies of Advanced Therapies, University of Ferrara, Ferrara, Italy
| | - Mariasole Perrone
- Department of Medical Sciences, Section of Experimental Medicine, Laboratory for Technologies of Advanced Therapies, University of Ferrara, Ferrara, Italy
| | - Simone Patergnani
- Department of Medical Sciences, Section of Experimental Medicine, Laboratory for Technologies of Advanced Therapies, University of Ferrara, Ferrara, Italy
| | - Caterina Boncompagni
- Department of Medical Sciences, Section of Experimental Medicine, Laboratory for Technologies of Advanced Therapies, University of Ferrara, Ferrara, Italy
| | - Alessandra Bortoluzzi
- Rheumatology Unit, Department of Medical Sciences, Università degli Studi di Ferrara and Azienda Ospedaliero-Universitaria S. Anna, Cona, Italy
| | - Marcello Govoni
- Rheumatology Unit, Department of Medical Sciences, Università degli Studi di Ferrara and Azienda Ospedaliero-Universitaria S. Anna, Cona, Italy
| | - Carlotta Giorgi
- Department of Medical Sciences, Section of Experimental Medicine, Laboratory for Technologies of Advanced Therapies, University of Ferrara, Ferrara, Italy
| | - Stefano Alivernini
- Rheumatology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Paolo Pinton
- Department of Medical Sciences, Section of Experimental Medicine, Laboratory for Technologies of Advanced Therapies, University of Ferrara, Ferrara, Italy
| | - Carlo Alberto Scirè
- Rheumatology Unit, Department of Medical Sciences, Università degli Studi di Ferrara and Azienda Ospedaliero-Universitaria S. Anna, Cona, Italy
- Epidemiology Research Unit, Italian Society for Rheumatology, Milan, Italy
| |
Collapse
|
22
|
Treatment of reactive arthritis with biological agents: a review. Biosci Rep 2021; 40:222065. [PMID: 32039436 PMCID: PMC7033307 DOI: 10.1042/bsr20191927] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 02/02/2020] [Accepted: 02/05/2020] [Indexed: 12/31/2022] Open
Abstract
The pathogenesis of reactive arthritis (ReA) has not been fully elucidated. In recent years, many researchers have confirmed that multiple cytokines are involved in the occurrence and development of ReA. Although ReA is self-limiting, it is still incurable for some patients who have no or a weak response to traditional drugs, such as non-steroidal anti-inflammatory agents, glucocorticoids and immunosuppressive agents. This is called refractory reactive arthritis. Currently, there is insufficient evidences for the treatment of refractory ReA with biological agents, though biological agents against cytokines have been developed over the past few years. This review summarizes the current development of clinical treatments of ReA with biological agents, which provides future investigations on refractory ReA with more evidence and references.
Collapse
|
23
|
Fiechter RH, de Jong HM, van Mens LJJ, Fluri IA, Tas SW, Baeten DLP, Yeremenko NG, van de Sande MGH. IL-12p40/IL-23p40 Blockade With Ustekinumab Decreases the Synovial Inflammatory Infiltrate Through Modulation of Multiple Signaling Pathways Including MAPK-ERK and Wnt. Front Immunol 2021; 12:611656. [PMID: 33746955 PMCID: PMC7971179 DOI: 10.3389/fimmu.2021.611656] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/08/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Psoriatic arthritis (PsA) is a chronic inflammatory joint disease within the spondyloarthritis spectrum. IL-12p40/IL-23p40 blockade reduces PsA disease activity, but its impact on synovial inflammation remains unclear. Objectives: To investigate the cellular and molecular pathways affected by IL-12p40/IL-23p40 blockade with ustekinumab in the synovium of PsA patients. Methods: Eleven PsA patients with at least one inflamed knee or ankle joint were included in a 24-week single-center open-label study and received ustekinumab 45 mg/sc according to standard care at week 0, 4, and 16. Besides clinical outcomes, synovial tissue (ST) samples were obtained by needle arthroscopy from an inflamed knee or ankle joint at baseline, week 12 and 24 and analyzed by immunohistochemistry, RNA-sequencing and real-time quantitative polymerase chain reaction (qPCR). Results: We obtained paired baseline and week 12, and paired baseline, week 12 and 24 ST samples from nine and six patients, respectively. Eight patients completed 24 weeks of clinical follow-up. At 12 weeks 6/11 patients met ACR20, 2/11 met ACR50 and 1/11 met ACR70 improvement criteria, at 24 weeks this was 3/8, 2/8 and 1/8 patients, respectively. Clinical and serological markers improved significantly. No serious adverse events occurred. We observed numerical decreases of all infiltrating cell subtypes at week 12, reaching statistical significance for CD68+ sublining macrophages. For some cell types this was even more pronounced at week 24, but clearly synovial inflammation was incompletely resolved. IL-17A and F, TNF, IL-6, IL-8, and IL-12p40 were not significantly downregulated in qPCR analysis of W12 total biopsies, only MMP3 and IL-23p19 were significantly decreased. RNA-seq analysis revealed 178 significantly differentially expressed genes between baseline and 12 weeks (FDR 0.1). Gene Ontology and KEGG terms enrichment analyses identified overrepresentation of biological processes as response to reactive oxygen species, chemotaxis, migration and angiogenesis as well as MAPK-ERK and PI3K-Akt signaling pathways among the downregulated genes and of Wnt signaling pathway among the upregulated genes. Furthermore, ACR20 responders and non-responders differed strikingly in gene expression profiles in a post-hoc exploratory analysis. Conclusions: Ustekinumab suppresses PsA synovial inflammation through modulation of multiple signal transduction pathways, including MAPK-ERK, Wnt and potentially PI3K-Akt signaling rather than by directly impacting the IL-17 pathway.
Collapse
Affiliation(s)
- Renée H Fiechter
- Amsterdam UMC, Department of Rheumatology and Clinical Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam UMC, Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Rheumatology and Immunology Center, Amsterdam, Netherlands
| | - Henriëtte M de Jong
- Amsterdam UMC, Department of Rheumatology and Clinical Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam UMC, Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Rheumatology and Immunology Center, Amsterdam, Netherlands
| | - Leonieke J J van Mens
- Amsterdam UMC, Department of Rheumatology and Clinical Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam UMC, Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Rheumatology and Immunology Center, Amsterdam, Netherlands
| | - Inka A Fluri
- Amsterdam UMC, Department of Rheumatology and Clinical Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam UMC, Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Rheumatology and Immunology Center, Amsterdam, Netherlands
| | - Sander W Tas
- Amsterdam UMC, Department of Rheumatology and Clinical Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam UMC, Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Rheumatology and Immunology Center, Amsterdam, Netherlands
| | - Dominique L P Baeten
- Amsterdam UMC, Department of Rheumatology and Clinical Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam UMC, Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Rheumatology and Immunology Center, Amsterdam, Netherlands
| | - Nataliya G Yeremenko
- Amsterdam UMC, Department of Rheumatology and Clinical Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam UMC, Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Rheumatology and Immunology Center, Amsterdam, Netherlands
| | - Marleen G H van de Sande
- Amsterdam UMC, Department of Rheumatology and Clinical Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam UMC, Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Rheumatology and Immunology Center, Amsterdam, Netherlands
| |
Collapse
|
24
|
Anti-IL-17A treatment reduces serum inflammatory, angiogenic and tissue remodeling biomarkers accompanied by less synovial high endothelial venules in peripheral spondyloarthritis. Sci Rep 2020; 10:21094. [PMID: 33273664 PMCID: PMC7713433 DOI: 10.1038/s41598-020-78204-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022] Open
Abstract
Spondyloarthritis (SpA) is characterized by inflammation and new bone formation. The exact pathophysiology underlying these processes remains elusive. We propose that the extensive neoangiogenesis in SpA could play a role both in sustaining/enhancing inflammation and in new bone formation. While ample data is available on effects of anti-TNF on angiogenesis, effects of IL-17A blockade on serum markers are largely unknown. We aimed to assess the impact of secukinumab (anti-IL-17A) on synovial neoangiogenesis in peripheral SpA, and how this related to changes in inflammatory and tissue remodeling biomarkers. Serum samples from 20 active peripheral SpA patients included in a 12 week open-label trial with secukinumab were analyzed for several markers of angiogenesis and tissue remodeling. Synovial biopsies taken before and after treatment were stained for vascular markers. Serum levels of MMP-3, osteopontin, IL-6 (all P < 0.001), IL-31, S100A8, S100A9, Vascular Endothelial Growth Factor A (VEGF-A), IL-33, TNF-α (all P < 0.05) decreased significantly upon anti-IL17A treatment. Secukinumab treatment resulted in a decrease in the number of synovial high endothelial venules and lymphoid aggregate score. These results indicate that anti-IL-17A not only diminishes inflammation, but also impacts angiogenesis and tissue remodeling/new bone formation. This may have important implications for disease progression and/or structural damage.
Collapse
|
25
|
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
|
26
|
Hospach T, Minden K, Huppertz HI. Reaktive Arthritis – ein Update. Monatsschr Kinderheilkd 2020. [DOI: 10.1007/s00112-020-01046-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
27
|
da Silva Cendon Duran C, Barreto Santiago M. Cutaneous Vasculitis During Secukinumab Treatment. Eur J Case Rep Intern Med 2020; 7:001815. [PMID: 33083361 DOI: 10.12890/2020_001815] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 06/18/2020] [Indexed: 11/05/2022] Open
Abstract
Secukinumab is an inhibitor of interleukin IL-17A, and is mainly used in the treatment of psoriasis, psoriatic arthritis and ankylosing spondylitis. Although rarely, this drug may induce paradoxical reactions, such as cutaneous vasculitis. Worldwide, only four previous cases of vasculitis induced by secukinumab have been reported. We herein report the first case described in Brazil of cutaneous vasculitis due to secukinumab in a patient with peripheral spondyloarthritis who demonstrated good resolution after withdrawal of the drug and initiation of etanercept. Clinicians should be aware of this rare but potentially serious adverse effect of secukinumab. LEARNING POINTS Treatment with biologics can cause vasculitis.The vasculitis may be independent of class of the biologic.Only five cases of vasculitis induced by secukinumab have been reported.
Collapse
Affiliation(s)
| | - Mittermayer Barreto Santiago
- Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Bahia, Brazil.,Serviços Especializados em Reumatologia da Bahia, Salvador, Bahia, Brazil.,Escola Bahiana de Medicina e Saúde Pública, Salvador, Bahia, Brazil
| |
Collapse
|
28
|
Abstract
Purpose of Review The aim of this paper is to provide an overview about reactive arthritis, with an update regarding pathophysiology and therapeutic approach of the disease, outlining the clinical features and diagnostic approach, based on recent literature review. Recent Findings Reactive arthritis is considered to be part of the spectrum of the spondyloarthritis. Its epidemiology is changing worldwide due to several reasons, among them are as follows: different diagnosis approach and clinical presentations, different grades of infection, microbiome changes, etc. The understanding of pathophysiological models is challenging, but recent studies contribute to elucidate the major factors involved in the development of the disease. The management of ReA depends on the triggering agent and the phase of disease, whether it is acute or chronic. Summary The association between the microbiome changes and spondyloarthropathies (ReA) is becoming increasingly evident. The results regarding the biologic treatment on refectory ReA are promising.
Collapse
|
29
|
Revisiting the gut-joint axis: links between gut inflammation and spondyloarthritis. Nat Rev Rheumatol 2020; 16:415-433. [PMID: 32661321 DOI: 10.1038/s41584-020-0454-9] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2020] [Indexed: 02/07/2023]
Abstract
Gut inflammation is strongly associated with spondyloarthritis (SpA), as exemplified by the high prevalence of inflammatory bowel disease (IBD) and the even higher occurrence of subclinical gut inflammation in patients with SpA. The gut-joint axis of inflammation in SpA is further reinforced by similarities in immunopathogenesis at both anatomical sites and by the clinical success of therapies blocking TNF and IL-23 in IBD and in some forms of SpA. Many genetic risk factors are shared between SpA and IBD, and changes in the composition of gut microbiota are seen in both diseases. Current dogma is that inflammation in SpA initiates in the gut and leads to joint inflammation; however, although conceptually attractive, some research does not support this causal relationship. For example, therapies targeting IL-17A are efficacious in the joint but not the gut, and interfering with gut trafficking by targeting molecules such as α4β7 in IBD can lead to onset or flares of SpA. Several important knowledge gaps remain that must be addressed in future studies. Determining the true nature of the gut-joint axis has real-world implications for the treatment of patients with co-incident IBD and SpA and for the repurposing of therapeutics from one disease to the other.
Collapse
|
30
|
|
31
|
Hile G, Kahlenberg JM, Gudjonsson JE. Recent genetic advances in innate immunity of psoriatic arthritis. Clin Immunol 2020; 214:108405. [PMID: 32247832 DOI: 10.1016/j.clim.2020.108405] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 03/28/2020] [Accepted: 03/28/2020] [Indexed: 12/19/2022]
Abstract
Psoriatic arthritis (PsA) is a heterogeneous disease that affects multiple organ systems including the peripheral and axial joints, entheses and nails. PsA is associated with significant comorbidities including cardiovascular, metabolic, and psychiatric diseases. The pathogenesis of PsA is complex and involves genetic, immunologic and environmental factors. Recent evidence suggests the heritability for PsA to be stronger and distinct from that of PsC. Prominent genes identified via GWAS for PsA include HLA-B/C, HLAB, IL12B, IL23R, TNP1, TRAF3IP3, and REL. We review the genetics of psoriatic arthritis and discuss the role of the innate immune system as important in the pathogenesis of PsA by focusing on key signaling pathways and cellular makeup. Understanding the candidate genes identified in PsA highlights pathways of critical importance to the pathogenesis of psoriatic disease including the key role of the innate immune response, mediated through IL-23/IL-17 axis, RANK and NFκB signaling pathways.
Collapse
Affiliation(s)
- Grace Hile
- Department of Dermatology, University of Michigan, Ann Arbor 48109, MI, USA.
| | - J Michelle Kahlenberg
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Johann E Gudjonsson
- Department of Dermatology, University of Michigan, Ann Arbor 48109, MI, USA.
| |
Collapse
|
32
|
Chen S, Paveley R, Kraal L, Sritharan L, Stevens E, Dedi N, Shock A, Shaw S, Juarez M, Yeremenko N, Baeten D, Payne A. Selective targeting of PI3Kδ suppresses human IL-17-producing T cells and innate-like lymphocytes and may be therapeutic for IL-17-mediated diseases. J Autoimmun 2020; 111:102435. [PMID: 32360069 DOI: 10.1016/j.jaut.2020.102435] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 03/01/2020] [Accepted: 03/02/2020] [Indexed: 12/19/2022]
Abstract
The delta isoform of phosphoinositide 3-kinase (PI3Kδ) regulates various lymphocyte functions. Considering the key pro-inflammatory role of IL-17A and IL-17F cytokines in psoriasis and spondyloarthritis (SpA), we investigated the potential of PI3Kδ blockade to suppress IL-17A, IL-17F and associated pro-inflammatory cytokines that could synergize with IL-17A and IL-17F. Using in vitro studies with primary human cells and ex vivo studies with inflamed target tissues, we assessed if seletalisib, a selective PI3Kδ inhibitor, suppresses cytokine production by T cells and innate-like lymphocytes, and if seletalisib modulates the inflammatory responses in stromal cell populations in psoriasis (human dermal fibroblasts (HDF)) and SpA (fibroblast-like synoviocytes (FLS)). In vitro, seletalisib inhibited the production of pro-inflammatory cytokines, including IL-17A and IL-17F, from peripheral blood mononuclear cells (PBMCs), T helper 17 (Th17) cells as well as γδ-T cells and mucosal-associated invariant T cells. This inhibition resulted in decreased inflammatory activation of HDF in co-culture systems. Seletalisib was also efficacious in inhibiting SpA PBMCs and synovial fluid mononuclear cells (SFMCs) from producing pro-inflammatory cytokines. Furthermore, supernatant derived from cultured seletalisib-treated Th17 cells showed reduced potency for activating inflammatory responses from cultured SpA FLS and decreased their osteogenic differentiation capacity. Finally, analysis of inflamed SpA synovial tissue biopsies revealed activation of the PI3K-Akt-mTOR pathway. We observed that ex vivo seletalisib treatment of inflamed synovial tissue reduced IL-17A and IL-17F expression. Collectively, inhibition of PI3Kδ reduces the production of pro-inflammatory cytokines from IL-17-producing adaptive and innate-like lymphocytes and thereby inhibits downstream inflammatory and tissue remodeling responses. PI3Kδ-targeting may therefore represent a novel therapeutic avenue for the treatment of IL-17-mediated chronic inflammatory diseases such as psoriasis and SpA.
Collapse
Affiliation(s)
- Sijia Chen
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology & immunology Center (ARC), Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, the Netherlands; Department of Experimental Immunology, Infection and Immunity Institute, Amsterdam University Medical Centers, location AMC, University of Amsterdam, the Netherlands.
| | | | - Lianne Kraal
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology & immunology Center (ARC), Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, the Netherlands; Department of Experimental Immunology, Infection and Immunity Institute, Amsterdam University Medical Centers, location AMC, University of Amsterdam, the Netherlands
| | - Lathees Sritharan
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology & immunology Center (ARC), Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, the Netherlands; Department of Experimental Immunology, Infection and Immunity Institute, Amsterdam University Medical Centers, location AMC, University of Amsterdam, the Netherlands
| | | | | | | | | | | | - Nataliya Yeremenko
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology & immunology Center (ARC), Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, the Netherlands; Department of Experimental Immunology, Infection and Immunity Institute, Amsterdam University Medical Centers, location AMC, University of Amsterdam, the Netherlands
| | - Dominique Baeten
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology & immunology Center (ARC), Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, the Netherlands; UCB Pharma, Slough, UK
| | | |
Collapse
|
33
|
Chen S, van Tok MN, Knaup VL, Kraal L, Pots D, Bartels L, Gravallese EM, Taurog JD, van de Sande M, van Duivenvoorde LM, Baeten DL. mTOR Blockade by Rapamycin in Spondyloarthritis: Impact on Inflammation and New Bone Formation in vitro and in vivo. Front Immunol 2020; 10:2344. [PMID: 32194539 PMCID: PMC7065603 DOI: 10.3389/fimmu.2019.02344] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 09/17/2019] [Indexed: 12/16/2022] Open
Abstract
Introduction: Spondyloarthritis (SpA) is characterized by inflammation, articular bone erosions and pathologic new bone formation. Targeting TNFα or IL-17A with current available therapies reduces inflammation in SpA, however, treatment of the bone pathology in SpA remains an unmet clinical need. Activation of the mammalian target Of rapamycin (mTOR) promotes IL-17A expression and osteogenesis. Therefore, the inhibition of mTOR (with rapamycin) could be a promising therapeutic avenue in SpA. Objectives: To investigate the effect of blocking mTOR on inflammation, bone erosions and new bone formation in SpA. Methods: Peripheral blood mononuclear cells (PBMCs) from patients with SpA were stimulated with anti-CD3/CD28 in the presence or absence of rapamycin and the resulting cytokine expression was assessed. Fibroblast-like synoviocytes (FLS) from SpA patients were assessed for osteogenic differentiation potential in conditions with TNFα, IL-17A, or TNFα plus IL-17A, in the presence or absence of rapamycin. HLA-B27/Huβ2m transgenic rats were immunized with low dose heat-inactivated Mycobacterium tuberculosis (M. tub), treated with 1.5 mg/kg rapamycin prophylactically or therapeutically and monitored for arthritis and spondylitis. Histology and mRNA analysis were performed after 5 weeks of treatment to assess inflammation and bone pathology. Results:In vitro TNFα and IL-17A protein production by SpA PBMCs was inhibited in the presence of rapamycin. Rapamycin also inhibited osteogenic differentiation of human SpA FLS. Ex vivo analysis of SpA synovial biopsies indicated activation of the mTOR pathway in the synovial tissue of SpA patients. In vivo, prophylactic treatment of HLA-B27/Huβ2m transgenic rats with rapamycin significantly inhibited the development and severity of inflammation in peripheral joints and spine (arthritis and spondylitis), with histological evidence of reduced bone erosions and new bone formation around peripheral joints. In addition, therapeutic treatment with rapamycin significantly decreased severity of arthritis and spondylitis, with peripheral joint histology showing reduced inflammation, bone erosions and new bone formation. IL-17A mRNA expression was decreased in the metacarpophalangeal joints after rapamycin treatment. Conclusion: mTOR blockade inhibits IL-17A and TNFα production by PBMCs, and osteogenic differentiation of FLS from patients with SpA in vitro. In the HLA-B27 transgenic rat model of SpA, rapamycin inhibits arthritis and spondylitis development and severity, reduces articular bone erosions, decreases pathologic new bone formation and suppresses IL-17A expression. These results may support efforts to evaluate the efficacy of targeting the mTOR pathway in SpA patients.
Collapse
Affiliation(s)
- Sijia Chen
- Department of Experimental Immunology, Infection and Immunity Institute, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Melissa N van Tok
- Department of Experimental Immunology, Infection and Immunity Institute, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Véronique L Knaup
- Department of Experimental Immunology, Infection and Immunity Institute, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Lianne Kraal
- Department of Experimental Immunology, Infection and Immunity Institute, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Désiree Pots
- Department of Experimental Immunology, Infection and Immunity Institute, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Lina Bartels
- Department of Experimental Immunology, Infection and Immunity Institute, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Ellen M Gravallese
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Joel D Taurog
- Internal Medicine, Rheumatic Diseases Division, UT Southwestern Medical Center, Dallas, TX, United States
| | - Marleen van de Sande
- Department of Experimental Immunology, Infection and Immunity Institute, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Leonie M van Duivenvoorde
- Department of Experimental Immunology, Infection and Immunity Institute, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Dominique L Baeten
- Department of Experimental Immunology, Infection and Immunity Institute, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,UCB Pharma, Slough, United Kingdom
| |
Collapse
|
34
|
Gonçalves dos Santos G, Delay L, Yaksh TL, Corr M. Neuraxial Cytokines in Pain States. Front Immunol 2020; 10:3061. [PMID: 32047493 PMCID: PMC6997465 DOI: 10.3389/fimmu.2019.03061] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/16/2019] [Indexed: 12/11/2022] Open
Abstract
A high-intensity potentially tissue-injuring stimulus generates a homotopic response to escape the stimulus and is associated with an affective phenotype considered to represent pain. In the face of tissue or nerve injury, the afferent encoding systems display robust changes in the input-output function, leading to an ongoing sensation reported as painful and sensitization of the nociceptors such that an enhanced pain state is reported for a given somatic or visceral stimulus. Our understanding of the mechanisms underlying this non-linear processing of nociceptive stimuli has led to our appreciation of the role played by the functional interactions of neural and immune signaling systems in pain phenotypes. In pathological states, neural systems interact with the immune system through the actions of a variety of soluble mediators, including cytokines. Cytokines are recognized as important mediators of inflammatory and neuropathic pain, supporting system sensitization and the development of a persistent pathologic pain. Cytokines can induce a facilitation of nociceptive processing at all levels of the neuraxis including supraspinal centers where nociceptive input evokes an affective component of the pain state. We review here several key proinflammatory and anti-inflammatory cytokines/chemokines and explore their underlying actions at four levels of neuronal organization: (1) peripheral nociceptor termini; (2) dorsal root ganglia; (3) spinal cord; and (4) supraspinal areas. Thus, current thinking suggests that cytokines by this action throughout the neuraxis play key roles in the induction of pain and the maintenance of the facilitated states of pain behavior generated by tissue injury/inflammation and nerve injury.
Collapse
Affiliation(s)
| | - Lauriane Delay
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA, United States
| | - Tony L. Yaksh
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA, United States
| | - Maripat Corr
- Division of Rheumatology, Allergy and Immunology, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
35
|
McGeachy MJ, Cua DJ, Gaffen SL. The IL-17 Family of Cytokines in Health and Disease. Immunity 2019; 50:892-906. [PMID: 30995505 DOI: 10.1016/j.immuni.2019.03.021] [Citation(s) in RCA: 809] [Impact Index Per Article: 161.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 03/14/2019] [Accepted: 03/21/2019] [Indexed: 12/20/2022]
Abstract
The interleukin 17 (IL-17) family of cytokines contains 6 structurally related cytokines, IL-17A through IL-17F. IL-17A, the prototypical member of this family, just passed the 25th anniversary of its discovery. Although less is known about IL-17B-F, IL-17A (commonly known as IL-17) has received much attention for its pro-inflammatory role in autoimmune disease. Over the past decade, however, it has become clear that the functions of IL-17 are far more nuanced than simply turning on inflammation. Accumulating evidence indicates that IL-17 has important context- and tissue-dependent roles in maintaining health during response to injury, physiological stress, and infection. Here, we discuss the functions of the IL-17 family, with a focus on the balance between the pathogenic and protective roles of IL-17 in cancer and autoimmune disease, including results of therapeutic blockade and novel aspects of IL-17 signal transduction regulation.
Collapse
Affiliation(s)
- Mandy J McGeachy
- Division of Rheumatology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| | | | - Sarah L Gaffen
- Division of Rheumatology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
36
|
Affiliation(s)
- Dae Hyun Yoo
- Department of Rheumatology, Hospital for Rheumatic Diseases, College of Medicine, Hanyang University, Seoul, Korea
| |
Collapse
|
37
|
Bianchi E, Rogge L. The IL-23/IL-17 pathway in human chronic inflammatory diseases – new insight from genetics and targeted therapies. Microbes Infect 2019; 21:246-253. [DOI: 10.1016/j.micinf.2019.06.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 04/09/2019] [Indexed: 02/06/2023]
|
38
|
The IL-23/IL-17 pathway in human chronic inflammatory diseases-new insight from genetics and targeted therapies. Genes Immun 2019; 20:415-425. [PMID: 31000797 DOI: 10.1038/s41435-019-0067-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/08/2019] [Accepted: 04/09/2019] [Indexed: 12/15/2022]
Abstract
Chronic inflammatory diseases such as rheumatoid arthritis, inflammatory bowel disease, spondyloarthritis, and psoriasis cause significant morbidity and are a considerable burden for the patients in terms of pain, impaired function, and diminished quality of life, as well as for society, because of the associated high health-care costs and loss of productivity. Our limited understanding of the pathogenic mechanisms involved in these diseases currently hinders early diagnosis and the development of more specific and effective therapies. The past years have been marked by considerable progress in our insight of the genetic basis of many diseases. In particular, genome-wide association studies (GWAS) performed with thousands of patients have provided detailed information about the genetic variants associated with a large number of chronic inflammatory diseases. These studies have brought to the forefront many genes linked to signaling pathways that were not previously known to be involved in pathogenesis, pointing to new directions in the study of disease mechanisms. GWAS also provided fundamental evidence for a key role of the immune system in the pathogenesis of these diseases, because many of the identified loci map to genes involved in different immune processes. However, the mechanisms by which disease-associated genetic variants act on disease development and the targeted cell populations remain poorly understood. The challenge of the post-GWAS era is to understand how these variants affect pathogenesis, to allow translation of genetic data into better diagnostics and innovative treatment strategies. Here, we review recent results that document the importance of the IL-23/IL-17 pathway for the pathogenesis of several chronic inflammatory diseases and summarize data that demonstrate how therapeutic targeting of this pathway can benefit affected patients.
Collapse
|
39
|
Menegatti S, Bianchi E, Rogge L. Anti-TNF Therapy in Spondyloarthritis and Related Diseases, Impact on the Immune System and Prediction of Treatment Responses. Front Immunol 2019; 10:382. [PMID: 30941119 PMCID: PMC6434926 DOI: 10.3389/fimmu.2019.00382] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 02/14/2019] [Indexed: 12/14/2022] Open
Abstract
Immune-mediated inflammatory diseases (IMIDs), such as spondyloarthritis (SpA), psoriasis, Crohn's disease (CD), and rheumatoid arthritis (RA) remain challenging illnesses. They often strike at a young age and cause lifelong morbidity, representing a considerable burden for the affected individuals and society. Pioneering studies have revealed the presence of a TNF-dependent proinflammatory cytokine cascade in several IMIDs, and the introduction of anti-TNF therapy 20 years ago has proven effective to reduce inflammation and clinical symptoms in RA, SpA, and other IMID, providing unprecedented clinical benefits and a valid alternative in case of failure or intolerable adverse effects of conventional disease-modifying antirheumatic drugs (DMARDs, for RA) or non-steroidal anti-inflammatory drugs (NSAIDs, for SpA). However, our understanding of how TNF inhibitors (TNFi) affect the immune system in patients is limited. This question is relevant because anti-TNF therapy has been associated with infectious complications. Furthermore, clinical efficacy of TNFi is limited by a high rate of non-responsiveness (30–40%) in RA, SpA, and other IMID, exposing a substantial fraction of patients to side-effects without clinical benefit. Despite the extensive use of TNFi, it is still not possible to determine which patients will respond to TNFi before treatment initiation. The recent introduction of antibodies blocking IL-17 has expanded the therapeutic options for SpA, as well as psoriasis and psoriatic arthritis. It is therefore essential to develop tools to guide treatment decisions for patients affected by SpA and other IMID, both to optimize clinical care and contain health care costs. After a brief overview of the biology of TNF, its receptors and currently used TNFi in the clinics, we summarize the progress that has been made to increase our understanding of the action of TNFi on the immune system in patients. We then summarize efforts dedicated to identify biomarkers that can predict treatment responses to TNFi and we conclude with a section dedicated to the recently introduced inhibitors of IL-17A and IL-23 in SpA and related diseases. The focus of this review is on SpA, however, we also refer to RA on topics for which only limited information is available on SpA in the literature.
Collapse
Affiliation(s)
- Silvia Menegatti
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Paris, France.,Unité Mixte de Recherche, Institut Pasteur/AP-HP Hôpital Cochin, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Elisabetta Bianchi
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Paris, France.,Unité Mixte de Recherche, Institut Pasteur/AP-HP Hôpital Cochin, Paris, France
| | - Lars Rogge
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Paris, France.,Unité Mixte de Recherche, Institut Pasteur/AP-HP Hôpital Cochin, Paris, France
| |
Collapse
|
40
|
Wendling D, Verhoeven F, Prati C. Anti-IL-17 monoclonal antibodies for the treatment of ankylosing spondylitis. Expert Opin Biol Ther 2018; 19:55-64. [DOI: 10.1080/14712598.2019.1554053] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Daniel Wendling
- Department of Rheumatology, CHRU de BESANCON, University Teaching Hospital, Besançon, France
- Université Bourgogne Franche-Comté, Besançon, France
| | - Frank Verhoeven
- Department of Rheumatology, CHRU de BESANCON, University Teaching Hospital, Besançon, France
| | - Clément Prati
- Department of Rheumatology, CHRU de BESANCON, University Teaching Hospital, Besançon, France
- Université Bourgogne Franche-Comté, Besançon, France
| |
Collapse
|