1
|
Ghouri M, Siddiqui NN, Lateef M, Avesi L, Khan R, Ghauri H, Asif E, Zehra S. Modified expression of JAK-STAT pathway genes in an in vivo rheumatoid arthritis model: A preclinical study to explore genetic insights. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167780. [PMID: 40073707 DOI: 10.1016/j.bbadis.2025.167780] [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: 11/06/2024] [Revised: 03/01/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic inflammatory disease characterised by inflammatory synovial tissue, joint deterioration, and effects on systems other than the joints. The biological process underlying the progression of the disease remains unknown, however cell-mediated immunity plays an important part in the onset of RA. The current study investigated the involvement of the JAK-STAT pathway genes (JAK-1, IL-6, and SOCS-2) in the pathogenesis of RA (Rheumatoid arthritis). METHODOLOGY The study was carried out on thirty male Albino Wistar rats categorised in to the three groups. The AIA (Adjuvant induced animal) model was utilised to study the disease pathogenesis. The haematoxylin and Eosin (H and E) was performed followed by ELISA and expression analyses by RT-q-PCR. The obtained data was analysed using one-way ANOVA (Analysis of Variance). RESULTS Histopathology confirmed that diseased group appeared to be severely impaired, demonstrating manifestations of inflammation with chronic as well as cartilage degenerative changes. Furthermore, chronic inflammation was also noticed in the intertrabecular area. The significant increased levels of JAK1, IL-6 and TYK-2 were recorded among RA group. The gene expression assessment indicated that higher expression of JAK-1 and IL-6 was linked to the further development of RA in the disease group. The SOSC2 (a negative regulator of the JAK-STAT pathway) was significantly (p < 0.01) downregulated. Moreover, SOCS2 may be unable to suppress the transcription of the related JAKs (IL-6 and JAK-1), resulting in the constant release of immune mediators and contributing to the pathophysiology of RA. CONCLUSIONS The JAK-STAT pathway may serve as the target for diagnosing and treating inflammatory and autoimmune disorders (RA). The findings may enhance therapeutic possibilities by investigating the possible implications of JAK-STAT pathway genes as candidates for progressive rheumatoid arthritis therapies.
Collapse
Affiliation(s)
- Maham Ghouri
- Karachi Institute of Biotechnology and Genetic Engineering (KIBGE), University of Karachi, Karachi, Sindh, Pakistan.
| | - Nadir Naveed Siddiqui
- Karachi Institute of Biotechnology and Genetic Engineering (KIBGE), University of Karachi, Karachi, Sindh, Pakistan.
| | - Mehreen Lateef
- Bahria University Medical and Dental College (BUMDC), Karachi, Sindh, Pakistan
| | - Lubna Avesi
- Department of Pathology, Dow University of Health Sciences, Karachi, Sindh, Pakistan.
| | - Rizma Khan
- Department of Biochemistry, Ziauddin University, Clifton, Karachi, Pakistan; Department of Molecular Genetics, Dr. Ziauddin Hospital, North Nazimabad, Karachi, Pakistan.
| | - Humaira Ghauri
- Bahria University Medical and Dental College (BUMDC), Karachi, Sindh, Pakistan.
| | - Ehtisham Asif
- Department of Biotechnology, University of Karachi, Karachi, Sindh, Pakistan
| | - Sitwat Zehra
- Karachi Institute of Biotechnology and Genetic Engineering (KIBGE), University of Karachi, Karachi, Sindh, Pakistan.
| |
Collapse
|
2
|
Ciccia F, McGonagle D, Thomas R, Marzo-Ortega H, Martin DA, Yndestad A, Volkov M. JAK inhibition and axial spondyloarthritis: new steps on the path to understanding pathophysiology. Front Immunol 2025; 16:1488357. [PMID: 40103808 PMCID: PMC11913702 DOI: 10.3389/fimmu.2025.1488357] [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: 08/29/2024] [Accepted: 02/13/2025] [Indexed: 03/20/2025] Open
Abstract
Axial spondyloarthritis (axSpA) is a chronic inflammatory disease that predominantly affects the sacroiliac joints and spine. Tumor necrosis factor (TNF) and interleukin (IL)-17A are key cytokines in disease pathogenesis and are established axSpA treatment targets. Recently, axSpA treatment options have been complemented by Janus kinase inhibitors (JAKi), which inhibit various cytokines without directly impacting TNF or IL-17 signaling. The effect of JAKi on axSpA remains under investigation: besides a JAK2-mediated (and potentially tyrosine kinase 2 [TYK2]-mediated) effect on the IL-23/IL-17 axis, emerging evidence suggests γδ T cells, type 3 innate lymphoid cells, and mucosa-associated invariant T cells, which are dependent on IL-7 and/or IL-15 and thus on JAK1, are strongly inhibited by JAKi used to treat axSpA. This review summarizes potential effects of JAKi on axSpA and shows evidence from pre-clinical/clinical studies. Greater understanding of the mechanisms of action of available treatments may improve knowledge of axSpA and pave the road for future therapies.
Collapse
Affiliation(s)
- Francesco Ciccia
- Dipartimento di Medicina di Precisione, Università della Campania L. Vanvitelli, Naples, Italy
| | - Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds and NIHR Leeds Biomedical Research Centre, The Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Ranjeny Thomas
- Frazer Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Helena Marzo-Ortega
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds and NIHR Leeds Biomedical Research Centre, The Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | | | | | | |
Collapse
|
3
|
Karjalainen A, Witalisz-Siepracka A, Prchal-Murphy M, Martin D, Sternberg F, Krunic M, Dolezal M, Fortelny N, Farlik M, Macho-Maschler S, Lassnig C, Meissl K, Amenitsch L, Lederer T, Pohl E, Gotthardt D, Bock C, Decker T, Strobl B, Müller M. Cell-type-specific requirement for TYK2 in murine immune cells under steady state and challenged conditions. Cell Mol Life Sci 2025; 82:98. [PMID: 40025196 PMCID: PMC11872851 DOI: 10.1007/s00018-025-05625-9] [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: 11/12/2024] [Revised: 01/31/2025] [Accepted: 02/17/2025] [Indexed: 03/04/2025]
Abstract
Tyrosine kinase 2 (TYK2) deficiency and loss or inhibition of kinase activity in men and mice leads to similar immune compromised phenotypes, predominantly through impairment of interferon (IFN) and interleukin 12 family responses. Here we relate the transcriptome changes to phenotypical changes observed in TYK2-deficient (Tyk2-/-) and TYK2 kinase-inactive (Tyk2K923E) mice in naïve splenic immune cells and upon ex vivo IFN treatment or in vivo tumor transplant infiltration. The TYK2 activities under homeostatic and both challenged conditions are highly cell-type-specific with respect to quantity and quality of transcriptionally dependent genes. The major impact of loss of TYK2 protein or kinase activity in splenic homeostatic macrophages, NK and CD8+ T cells and tumor-derived cytolytic cells is on IFN responses. While reportedly TYK2 deficiency leads to partial impairment of IFN-I responses, we identified cell-type-specific IFN-I-repressed gene sets completely dependent on TYK2 kinase activity. Reported kinase-inactive functions of TYK2 relate to signaling crosstalk, metabolic functions and cell differentiation or maturation. None of these phenotypes relates to respective enriched gene sets in the TYK2 kinase-inactive cell types. Nonetheless, the scaffolding functions of TYK2 are capable to change transcriptional activities at single gene levels and chromatin accessibility at promoter-distal regions upon cytokine treatment most prominently in CD8+ T cells. The cell-type-specific transcriptomic and epigenetic effects of TYK2 shed new light on the biology of this JAK family member and are relevant for current and future treatment of autoimmune and inflammatory diseases with TYK2 inhibitors.
Collapse
Affiliation(s)
- Anzhelika Karjalainen
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Agnieszka Witalisz-Siepracka
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
- Division Pharmacology, Karl Landsteiner University of Health Sciences, Krems an Der Donau, Austria
| | - Michaela Prchal-Murphy
- Pharmacology and Toxicology, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - David Martin
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Felix Sternberg
- Physiology and Biophysics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
- Department of Nutritional Sciences, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Milica Krunic
- Campus Tulln, University of Applied Sciences Wiener Neustadt, Wiener Neustadt, Austria
| | - Marlies Dolezal
- Platform Biostatistics and Bioinformatics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Nikolaus Fortelny
- Department of Biosciences and Medical Biology, Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, Austria
| | - Matthias Farlik
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Sabine Macho-Maschler
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Caroline Lassnig
- Core Facility VetBiomodels, University of Veterinary Medicine, Vienna, Austria
| | - Katrin Meissl
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Lena Amenitsch
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Therese Lederer
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Elena Pohl
- Physiology and Biophysics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Dagmar Gotthardt
- Division Pharmacology, Karl Landsteiner University of Health Sciences, Krems an Der Donau, Austria
| | - Christoph Bock
- Cemm Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Institute of Artificial Intelligence, Center for Medical Data Science, Medical University of Vienna, Vienna, Austria
| | - Thomas Decker
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Center for Molecular Biology, Department of Microbiology, Immunobiology and Genetics, University of Vienna, Vienna, Austria
| | - Birgit Strobl
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Mathias Müller
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria.
| |
Collapse
|
4
|
Navid F, Chen L, Bowness P, Colbert RA. HLA-B27 and spondyloarthritis: at the crossroads of innate and adaptive immunity. Nat Rev Rheumatol 2025; 21:77-87. [PMID: 39623156 DOI: 10.1038/s41584-024-01189-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2024] [Indexed: 01/29/2025]
Abstract
HLA-B*27 confers a strong risk of developing spondyloarthritis (SpA), which includes axial SpA with or without peripheral arthritis, enthesitis, acute anterior uveitis and gastrointestinal inflammation. Although no definitive mechanism has been established to explain the role of this HLA class I protein in the pathogenesis of SpA, three main hypotheses have emerged. First is the idea that self-peptides displayed by HLA-B27 resemble microorganism-derived peptides, leading to the expansion of autoreactive CD8+ T cells that trigger disease. The second and third hypotheses focus on aberrant properties of HLA-B27, including its tendency to form cell-surface dimers that can activate innate killer immunoglobulin-like receptors on CD4+ T helper 17 cells, triggering the production of pathogenic cytokines. HLA-B27 also misfolds in the endoplasmic reticulum, which can activate the unfolded protein response, increasing IL-23 expression and thereby promoting the production of type 17 cytokines. HLA-B27 misfolding in mesenchymal stem cells has also been linked to enhanced bone formation by mesenchymal stem cell-derived osteoblasts, which could contribute to structural damage in axial SpA. In this Review we summarize prevailing ideas about the role of HLA-B27 in SpA, discuss the latest developments as well as the gaps in current knowledge, and provide recommendations for future research to address these unmet needs.
Collapse
Affiliation(s)
- Fatemeh Navid
- Pediatric Translational Research Branch, National Institute of Arthritis, Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Liye Chen
- Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, Oxford University, Oxford, UK
| | - Paul Bowness
- Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, Oxford University, Oxford, UK
| | - Robert A Colbert
- Pediatric Translational Research Branch, National Institute of Arthritis, Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
5
|
Wei Y, Zhang S, Shao F, Sun Y. Ankylosing spondylitis: From pathogenesis to therapy. Int Immunopharmacol 2025; 145:113709. [PMID: 39644789 DOI: 10.1016/j.intimp.2024.113709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/22/2024] [Accepted: 11/22/2024] [Indexed: 12/09/2024]
Abstract
Ankylosing spondylitis (AS) is an autoimmune rheumatic disease that primarily affects the axial joints, with its etiology complex and still not fully understood. The unknown pathogenesis of AS limits the development of treatment strategies, so keeping up-to-date with the current research on AS can help in searching for potential therapeutic targets. In addition to the classic HLA-B27 genetic susceptibility and Th17-related inflammatory signals, increasing research is focusing on the influence of autoantigen-centered autoimmune responses and bone stromal cells on the onset of AS. Autoantigens derived from gut microbiota and preferential TCR both exacerbate the autoimmune response in patients with AS. Furthermore, dysregulated bone metabolism also promotes pathological new bone formation in AS. Current treatments approved for AS almost focus on the management of inflammation with inconsistent treatment results due to the heterogeneity of patients. In this review, we systematically summarized various pathogenesis and management of AS, meanwhile discussed the underlying risk factors and potential therapeutic targets.
Collapse
Affiliation(s)
- Yuxiao Wei
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, Jiangsu, China.
| | - Shuqiong Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, Jiangsu, China.
| | - Fenli Shao
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, Jiangsu, China.
| |
Collapse
|
6
|
Jaguan D, Nguyen KT, Goldfarb N. Selective small molecule inhibitors for hidradenitis suppurativa: Today and tomorrow. J Am Acad Dermatol 2024; 91:S31-S36. [PMID: 39626997 DOI: 10.1016/j.jaad.2024.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 12/12/2024]
Abstract
Hidradenitis suppurativa (HS) is an autoinflammatory condition characterized by abscesses, inflammatory nodules, and tunnels in intertriginous sites of the body. The pathogenesis of HS involves follicular occlusion in combination with environmental, genetic, hormonal, and metabolic factors. HS lesions are characterized by an influx of neutrophils, histiocytes, B and T cells, and upregulation of proinflammatory cytokines such as tumor necrosis factor-α, interleukin-1, interleukin-17, and interferons. Selective small molecule inhibitors (SMIs) are organic compounds that bind to active sites on target proteins involved in inflammatory signaling pathways, most commonly blocking enzymes, ion channels and receptors. SMIs are divided into conventional and selective SMIs. Selective SMIs are further subdivided into kinase and nonkinase SMIs. Currently there are five selective SMIs available in the United States with demonstrated efficacy for HS in clinical studies including apremilast, topical ruxolitinib, upadacitinib, fostamatinib, and sirolimus. These selective SMIs target four pathways hypothesized to be important to HS pathogenesis including phosphodiestase 4, Janus kinases, spleen tyrosine kinase, and mammalian target of rapamycin. Several new SMIs are currently in the clinical trial pipeline targeting Bruton's tyrosine kinase, aryl hydrocarbon receptors, heat shock protein 90 as well as interleukin-1 and -17 signaling pathways.
Collapse
Affiliation(s)
- Daniella Jaguan
- Georgetown University School of Medicine, Washington, District of Columbia; Department of Dermatology, University of Minnesota, Minneapolis, Minnesota
| | - Kim T Nguyen
- Department of Dermatology, University of Minnesota, Minneapolis, Minnesota; University of Nebraska Medical Center, College of Medicine, Omaha, Nebraska
| | - Noah Goldfarb
- Department of Dermatology, University of Minnesota, Minneapolis, Minnesota; Department of Medicine, University of Minnesota, Minneapolis, Minnesota; Departments of Medicine and Dermatology, Minneapolis VA Health Care System, Minneapolis, Minnesota.
| |
Collapse
|
7
|
Lai Y, Wu X, Jiang Z, Fang Y, Liu X, Hong D, Jiang Y, Tan G, Tang S, Lu S, Wei D, Hwang ST, Lam KS, Wang L, Huang Y, Shi Z. Topical treatment of tyrosine kinase 2 inhibitor through borneol-embedded hydrogel: Evaluation for preventive, therapeutic, and Recurrent management of psoriasis. Bioact Mater 2024; 41:83-95. [PMID: 39104775 PMCID: PMC11298611 DOI: 10.1016/j.bioactmat.2024.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 08/07/2024] Open
Abstract
Psoriasis, an immune-mediated inflammatory skin disorder characterized by a chronically relapsing-remitting course, continues to be primarily managed through topical therapy. While oral administration of tyrosine kinase 2 inhibitors (TYK2i) stands as an effective approach for psoriasis treatment, the potential efficacy of topical application of TYK2i remains unexplored. Herein, the carbomer/alginic acid hydrogel is embedded with borneol (BO) as a new topical carrier of TYK2i for achieving enhanced transdermal permeation and anti-psoriasis efficacy. The hydrogel system, i.e., TYK2i-BO-gel, exhibits significantly improved preventative and therapeutic effects in mice models of psoriasiform dermatitis, as evidenced by phenotypical images, psoriasis severity score index (PSI), histology, immunohistochemical staining, and PCR analysis. Remarkably, TYK2i-BO-gel outperforms conventional topical corticosteroid therapy by significantly preventing psoriatic lesion recurrence as measured by a nearly 50 % reduction in ear thickness changes (p < 0.0001), PSI (p < 0.0001) and epidermal thickness (p < 0.05). Moreover, a strengthened anti-inflammatory effect caused by TYK2i-BO-gel is seen in a human skin explant model, implying its potential application for human patients. With the addition of BO, the TYK2i-BO-gel not only increases skin permeability but also inhibits the expression of antimicrobial peptides in keratinocytes and facilitates the anti-Th17 response of TYK2i with suppressed activation of STAT3. Therefore, this work represents the accessibility and effectiveness of TYK2i-BO-hydrogel as a new topical formulation for anti-psoriasis management and shows great potential for clinical application.
Collapse
Affiliation(s)
- Yuhsien Lai
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 510120, China
| | - Xuesong Wu
- Department of Dermatology, University of California-Davis, Sacramento, CA, 95817, USA
| | - Zhuoyu Jiang
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 510120, China
| | - Yifei Fang
- School of Biomedical Engineering, Sun Yat-sen University, 518107, China
| | - Xiuting Liu
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 510120, China
| | - Dan Hong
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 510120, China
| | - Yanyun Jiang
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 510120, China
| | - Guozhen Tan
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 510120, China
| | - Shiqi Tang
- Department of Biochemistry and Molecular Medicine, University of California-Davis, Sacramento, CA, 95817, USA
| | - Siyao Lu
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 510120, China
| | - David Wei
- Department of Dermatology, University of California-Davis, Sacramento, CA, 95817, USA
| | - Sam T. Hwang
- Department of Dermatology, University of California-Davis, Sacramento, CA, 95817, USA
| | - Kit S. Lam
- Department of Biochemistry and Molecular Medicine, University of California-Davis, Sacramento, CA, 95817, USA
| | - Liangchun Wang
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 510120, China
| | - Yanyu Huang
- Department of Biochemistry and Molecular Medicine, University of California-Davis, Sacramento, CA, 95817, USA
| | - Zhenrui Shi
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 510120, China
| |
Collapse
|
8
|
Ramakrishna C, Mason A, Edwards CJ. Tyrosine kinase 2 inhibitors in autoimmune diseases. Autoimmun Rev 2024; 23:103649. [PMID: 39349269 DOI: 10.1016/j.autrev.2024.103649] [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: 07/22/2024] [Revised: 09/27/2024] [Accepted: 09/27/2024] [Indexed: 10/02/2024]
Abstract
Tyk2 is a member of the JAK kinase family. It is an important mediator in pro-inflammatory signalling, implicated in both innate and adaptive immune system. Activation of Tyk2 is believed to be integral to cellular processes that contribute to the development and progression of autoimmune disorders. Selective targeting of Tyk2 may reduce the number of adverse events as compared to non-selective JAK inhibitors. Therefore, in recent years there has been a growing body of research examining the inhibition of Tyk2 as a therapeutic intervention in autoimmune disease. Deucravacitinib has been approved for the treatment of moderate to severe skin psoriasis. This drug and other novel Tyk2 inhibitors are now being explored as therapies for multiple autoimmune diseases, including psoriatic arthritis, SLE, Sjogren's, dermatomyositis, inflammatory bowel disease, uveitis, hidradenitis suppurativa and others. Tyk2 inhibitors offer a potentially exciting new treatment option across a wide range of autoimmune diseases. We discuss Tyk2 inhibition, the current evidence for its usage to date, ongoing trials and what the future might hold.
Collapse
Affiliation(s)
- Chethana Ramakrishna
- Department of Rheumatology, University Hospital Southampton NHS Foundation Trust, Tremona Road, Southampton SO16 6YD, UK.
| | - Alice Mason
- Department of Rheumatology, University Hospital Southampton NHS Foundation Trust, Tremona Road, Southampton SO16 6YD, UK
| | - Christopher J Edwards
- Department of Rheumatology, University Hospital Southampton NHS Foundation Trust, Tremona Road, Southampton SO16 6YD, UK; NIHR Southampton clinical research facility, University Hospitals Southampton NHS Foundation Trust, Southampton, UK
| |
Collapse
|
9
|
Eshaq AM, Flanagan TW, Hassan SY, Al Asheikh SA, Al-Amoudi WA, Santourlidis S, Hassan SL, Alamodi MO, Bendhack ML, Alamodi MO, Haikel Y, Megahed M, Hassan M. Non-Receptor Tyrosine Kinases: Their Structure and Mechanistic Role in Tumor Progression and Resistance. Cancers (Basel) 2024; 16:2754. [PMID: 39123481 PMCID: PMC11311543 DOI: 10.3390/cancers16152754] [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: 05/29/2024] [Revised: 06/30/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Protein tyrosine kinases (PTKs) function as key molecules in the signaling pathways in addition to their impact as a therapeutic target for the treatment of many human diseases, including cancer. PTKs are characterized by their ability to phosphorylate serine, threonine, or tyrosine residues and can thereby rapidly and reversibly alter the function of their protein substrates in the form of significant changes in protein confirmation and affinity for their interaction with protein partners to drive cellular functions under normal and pathological conditions. PTKs are classified into two groups: one of which represents tyrosine kinases, while the other one includes the members of the serine/threonine kinases. The group of tyrosine kinases is subdivided into subgroups: one of them includes the member of receptor tyrosine kinases (RTKs), while the other subgroup includes the member of non-receptor tyrosine kinases (NRTKs). Both these kinase groups function as an "on" or "off" switch in many cellular functions. NRTKs are enzymes which are overexpressed and activated in many cancer types and regulate variable cellular functions in response to extracellular signaling-dependent mechanisms. NRTK-mediated different cellular functions are regulated by kinase-dependent and kinase-independent mechanisms either in the cytoplasm or in the nucleus. Thus, targeting NRTKs is of great interest to improve the treatment strategy of different tumor types. This review deals with the structure and mechanistic role of NRTKs in tumor progression and resistance and their importance as therapeutic targets in tumor therapy.
Collapse
Affiliation(s)
- Abdulaziz M. Eshaq
- Department of Epidemiology and Biostatistics, Milken Institute School of Public Health, George Washington University, Washington, DC 20052, USA;
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Thomas W. Flanagan
- Department of Pharmacology and Experimental Therapeutics, LSU Health Sciences Center, New Orleans, LA 70112, USA;
| | - Sofie-Yasmin Hassan
- Department of Pharmacy, Faculty of Science, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany;
| | - Sara A. Al Asheikh
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Waleed A. Al-Amoudi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Simeon Santourlidis
- Institute of Cell Therapeutics and Diagnostics, University Medical Center of Duesseldorf, 40225 Duesseldorf, Germany;
| | - Sarah-Lilly Hassan
- Department of Chemistry, Faculty of Science, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany;
| | - Maryam O. Alamodi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Marcelo L. Bendhack
- Department of Urology, Red Cross University Hospital, Positivo University, Rua Mauá 1111, Curitiba 80030-200, Brazil;
| | - Mohammed O. Alamodi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Youssef Haikel
- Institut National de la Santé et de la Recherche Médicale, University of Strasbourg, 67000 Strasbourg, France;
- Department of Operative Dentistry and Endodontics, Dental Faculty, 67000 Strasbourg, France
- Pôle de Médecine et Chirurgie Bucco-Dentaire, Hôpital Civil, Hôpitaux Universitaire de Strasbourg, 67000 Strasbourg, France
| | - Mossad Megahed
- Clinic of Dermatology, University Hospital of Aachen, 52074 Aachen, Germany;
| | - Mohamed Hassan
- Institut National de la Santé et de la Recherche Médicale, University of Strasbourg, 67000 Strasbourg, France;
- Department of Operative Dentistry and Endodontics, Dental Faculty, 67000 Strasbourg, France
- Research Laboratory of Surgery-Oncology, Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
10
|
Nakamura A, Jo S, Nakamura S, Aparnathi MK, Boroojeni SF, Korshko M, Park YS, Gupta H, Vijayan S, Rockel JS, Kapoor M, Jurisica I, Kim TH, Haroon N. HIF-1α and MIF enhance neutrophil-driven type 3 immunity and chondrogenesis in a murine spondyloarthritis model. Cell Mol Immunol 2024; 21:770-786. [PMID: 38839914 PMCID: PMC11214626 DOI: 10.1038/s41423-024-01183-5] [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: 10/29/2023] [Accepted: 05/08/2024] [Indexed: 06/07/2024] Open
Abstract
The hallmarks of spondyloarthritis (SpA) are type 3 immunity-driven inflammation and new bone formation (NBF). Macrophage migration inhibitory factor (MIF) was found to be a key driver of the pathogenesis of SpA by amplifying type 3 immunity, yet MIF-interacting molecules and networks remain elusive. Herein, we identified hypoxia-inducible factor-1 alpha (HIF1A) as an interacting partner molecule of MIF that drives SpA pathologies, including inflammation and NBF. HIF1A expression was increased in the joint tissues and synovial fluid of SpA patients and curdlan-injected SKG (curdlan-SKG) mice compared to the respective controls. Under hypoxic conditions in which HIF1A was stabilized, human and mouse neutrophils exhibited substantially increased expression of MIF and IL-23, an upstream type 3 immunity-related cytokine. Similar to MIF, systemic overexpression of IL-23 induced SpA pathology in SKG mice, while the injection of a HIF1A-selective inhibitor (PX-478) into curdlan-SKG mice prevented or attenuated SpA pathology, as indicated by a marked reduction in the expression of MIF and IL-23. Furthermore, genetic deletion of MIF or HIF1A inhibition with PX-478 in IL-23-overexpressing SKG mice did not induce evident arthritis or NBF, despite the presence of psoriasis-like dermatitis and blepharitis. We also found that MIF- and IL-23-expressing neutrophils infiltrated areas of the NBF in curdlan-SKG mice. These neutrophils potentially increased chondrogenesis and cell proliferation via the upregulation of STAT3 in periosteal cells and ligamental cells during endochondral ossification. Together, these results provide supporting evidence for an MIF/HIF1A regulatory network, and inhibition of HIF1A may be a novel therapeutic approach for SpA by suppressing type 3 immunity-mediated inflammation and NBF.
Collapse
Affiliation(s)
- Akihiro Nakamura
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, M5T 0S8, Canada.
- Krembil Research Institute, University Health Network, Toronto, ON, M5T 0S8, Canada.
- Institute of Medical Science, Temerty Faculty of Medicine of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada.
- Department of Medicine, Division of Rheumatology, Queen's University, Kingston, ON, K7L, 2V6, Canada.
- Translational Institute of Medicine, School of Medicine, Queen's University, Kingston, ON, K7L 2V6, Canada.
- Division of Rheumatology, Kingston Health Science Centre, Kingston, ON, K7L 2V6, Canada.
| | - Sungsin Jo
- Hanyang University Institute for Rheumatology Research (HYIRR), Seoul, 04763, Republic of Korea
- Department of Biology, College of Natural Sciences, Soonchunhyang University, Asan, 31538, Republic of Korea
| | - Sayaka Nakamura
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
| | - Mansi K Aparnathi
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
| | - Shaghayegh Foroozan Boroojeni
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Institute of Medical Science, Temerty Faculty of Medicine of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Mariia Korshko
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
| | - Ye-Soo Park
- Department of Orthopedic Surgery, Guri Hospital, Hanyang University College of Medicine, Guri, 11293, Republic of Korea
| | - Himanshi Gupta
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
| | - Sandra Vijayan
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
| | - Jason S Rockel
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
| | - Mohit Kapoor
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Department of Surgery and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5T 1P5, Canada
| | - Igor Jurisica
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Departments of Medical Biophysics and Comp. Science and Faculty of Dentistry, University of Toronto, Toronto, ON, M5G 1L7, Canada
- Institute of Neuroimmunology, Slovak Academy of Sciences, 85410, Bratislava, Slovakia
| | - Tae-Hwan Kim
- Hanyang University Institute for Rheumatology Research (HYIRR), Seoul, 04763, Republic of Korea
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, 04763, Republic of Korea
| | - Nigil Haroon
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, M5T 0S8, Canada.
- Krembil Research Institute, University Health Network, Toronto, ON, M5T 0S8, Canada.
- Institute of Medical Science, Temerty Faculty of Medicine of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
11
|
Raychaudhuri SP, Shah RJ, Banerjee S, Raychaudhuri SK. JAK-STAT Signaling and Beyond in the Pathogenesis of Spondyloarthritis and Their Clinical Significance. Curr Rheumatol Rep 2024; 26:204-213. [PMID: 38492148 PMCID: PMC11116266 DOI: 10.1007/s11926-024-01144-x] [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: 02/26/2024] [Indexed: 03/18/2024]
Abstract
PURPOSE OF REVIEW Janus kinase-signal transducers and activators of transcription cell signaling proteins (JAK-STATs) play a key regulatory role in functioning of several inflammatory cytokines. JAK-STAT signaling proteins are the key regulators of the cytokine/cytokine receptor system involved in the pathogenesis of various autoimmune disease including spondyloarthritis (SpA). This article mainly highlights the JAK-STAT signaling system, its association with the relevant cytokine/cytokine-receptor system, and its regulatory role in pathogenesis of SpA. Also, we have briefly addressed the principle for the use JAKi in SpA and the current status of use of JAK inhibitors (JAKi) in SpA. RECENT FINDINGS Recent developments with newer JAK molecules as well as other molecules beyond JAK inhibitors are now an exciting field for the development of novel therapies for autoimmune diseases and various malignant conditions. In this article, we have provided a special emphasis on how various cell signaling systems beyond JAK/STAT pathway are relevant to SpA and have provided a comprehensive review on this upcoming field in respect to the novel TYK2 inhibitors, RORγT inhibitors, mTOR inhibitors, NGF inhibitors, and various STAT kinase inhibitors. SpA are a group of autoimmune diseases with multifactorial etiologies. SpA is linked with genetic predisposition, environmental risk factors, and the immune system-mediated systemic inflammation. Here, we have provided the regulatory role of JAK/STAT pathway and other intracellular signaling system in the pathogenesis of SpA and its therapeutic relevance.
Collapse
Affiliation(s)
- Siba P Raychaudhuri
- Department of Rheumatology, UC Davis Medical Center, Sacramento, CA, USA.
- VA Sacramento Medical Center, Department of Veterans Affairs, Northern California Health Care System, Mather, CA, USA.
- UC Davis School of Medicine, Davis, CA, USA.
| | - Ruchi J Shah
- Department of Rheumatology, UC Davis Medical Center, Sacramento, CA, USA
| | - Sneha Banerjee
- VA Sacramento Medical Center, Department of Veterans Affairs, Northern California Health Care System, Mather, CA, USA
| | - Smriti K Raychaudhuri
- VA Sacramento Medical Center, Department of Veterans Affairs, Northern California Health Care System, Mather, CA, USA
| |
Collapse
|
12
|
Won EJ, Lee YJ, Kim MJ, Lee HI, Jang HH, Kim SH, Yoo HM, Cho N, Shim SC, Kim TJ. A potential role of protein extractions from Metagonimus yokogawai in amelionating inflammation in patients with ankylosing spondylitis. Exp Parasitol 2024; 259:108718. [PMID: 38369180 DOI: 10.1016/j.exppara.2024.108718] [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: 11/20/2023] [Revised: 02/04/2024] [Accepted: 02/08/2024] [Indexed: 02/20/2024]
Abstract
Helminth infections and their components has been recognized to have a positive impact on the immune system. This study aimed to investigate the potential of Metagonimus yokogawai-derived proteins (MYp) to provide protection against ankylosing spondylitis (AS) through modulation of immune responses. The cytotoxicity of MYp at various doses was first assessed using MTS and flow cytometry. Peripheral blood mononuclear cells (PBMCs) were collected from AS patients, and the production of inflammatory cytokines was analyzed through flow cytometry. In the experiments with SKG mice, MYp or vehicle was administered and inflammation was evaluated through immunohistochemistry and enzyme-linked immunosorbent assay. The results showed that MYp did not decrease cell viability of PBMCs even after 48 h. Additionally, the frequencies of IFN-γ and IL-17A producing cells were significantly reduced after MYp treatment in the PBMC cultures. Furthermore, MYp treatment significantly suppressed arthritis and enthesitis in the SKG mouse model. The results suggest the first evidence that MYp can effectively alleviate clinical symptoms and restore cytokine balance in patients with AS.
Collapse
Affiliation(s)
- Eun Jeong Won
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Yu Jeong Lee
- Department of Biomedical Sciences, Graduate School of Chonnam National University, Republic of Korea; Department of Rheumatology, Chonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Moon-Ju Kim
- Department of Rheumatology, Chonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Hae-In Lee
- Department of Rheumatology, Chonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Hyun Hee Jang
- Department of Biomedical Sciences, Graduate School of Chonnam National University, Republic of Korea; Department of Rheumatology, Chonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Seong Hoon Kim
- Department of Biomedical Sciences, Graduate School of Chonnam National University, Republic of Korea; Department of Rheumatology, Chonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Hee Min Yoo
- Microbiological Analysis Team, Biometrology Group, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Republic of Korea
| | - Namki Cho
- College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Seung Cheol Shim
- Division of Rheumatology, Daejeon Rheumatoid & Degenerative Arthritis Center, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Tae-Jong Kim
- Department of Biomedical Sciences, Graduate School of Chonnam National University, Republic of Korea; Department of Rheumatology, Chonnam National University Medical School and Hospital, Gwangju, Republic of Korea.
| |
Collapse
|
13
|
Mine K, Nagafuchi S, Akazawa S, Abiru N, Mori H, Kurisaki H, Shimoda K, Yoshikai Y, Takahashi H, Anzai K. TYK2 signaling promotes the development of autoreactive CD8 + cytotoxic T lymphocytes and type 1 diabetes. Nat Commun 2024; 15:1337. [PMID: 38351043 PMCID: PMC10864272 DOI: 10.1038/s41467-024-45573-9] [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: 08/16/2023] [Accepted: 01/25/2024] [Indexed: 02/16/2024] Open
Abstract
Tyrosine kinase 2 (TYK2), a member of the JAK family, has attracted attention as a potential therapeutic target for autoimmune diseases. However, the role of TYK2 in CD8+ T cells and autoimmune type 1 diabetes (T1D) is poorly understood. In this study, we generate Tyk2 gene knockout non-obese diabetes (NOD) mice and demonstrate that the loss of Tyk2 inhibits the development of autoreactive CD8+ T-BET+ cytotoxic T lymphocytes (CTLs) by impairing IL-12 signaling in CD8+ T cells and the CD8+ resident dendritic cell-driven cross-priming of CTLs in the pancreatic lymph node (PLN). Tyk2-deficient CTLs display reduced cytotoxicity. Increased inflammatory responses in β-cells with aging are dampened by Tyk2 deficiency. Furthermore, treatment with BMS-986165, a selective TYK2 inhibitor, inhibits the expansion of T-BET+ CTLs, inflammation in β-cells and the onset of autoimmune T1D in NOD mice. Thus, our study reveals the diverse roles of TYK2 in driving the pathogenesis of T1D.
Collapse
Affiliation(s)
- Keiichiro Mine
- Division of Metabolism and Endocrinology, Faculty of Medicine, Saga University, Saga, Japan.
- Division of Host Defense, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.
| | - Seiho Nagafuchi
- Division of Metabolism and Endocrinology, Faculty of Medicine, Saga University, Saga, Japan
| | - Satoru Akazawa
- Department of Endocrinology and Metabolism, Unit of Translational Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Norio Abiru
- Department of Endocrinology and Metabolism, Unit of Translational Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Midori Clinic, Nagasaki, Japan
| | - Hitoe Mori
- Division of Metabolism and Endocrinology, Faculty of Medicine, Saga University, Saga, Japan
| | - Hironori Kurisaki
- Department of Medical Science and Technology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuya Shimoda
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Yasunobu Yoshikai
- Division of Host Defense, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Hirokazu Takahashi
- Division of Metabolism and Endocrinology, Faculty of Medicine, Saga University, Saga, Japan
- Liver Center, Saga University Hospital, Saga University, Saga, Japan
| | - Keizo Anzai
- Division of Metabolism and Endocrinology, Faculty of Medicine, Saga University, Saga, Japan
| |
Collapse
|
14
|
Feng X, Qiao J, Xu W. Impact of immune regulation and differentiation dysfunction of mesenchymal stem cells on the disease process in ankylosing spondylitis and prospective analysis of stem cell transplantation therapy. Postgrad Med J 2023; 99:1138-1147. [PMID: 37689998 DOI: 10.1093/postmj/qgad073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/19/2023] [Accepted: 08/11/2023] [Indexed: 09/11/2023]
Abstract
Ankylosing spondylitis (AS) is a rheumatic bone and joint disease caused by inflammation, erosion, and pathological bone formation. The pathological features of chronic inflammation, bone destruction, and pathological ossification occur due to the disruption of the body's immune regulation and altered bone remodeling balance. Mesenchymal stem cells (MSCs) have multidirectional differentiation potential and immunomodulatory functions and play an important role in immune regulation and bone formation. The immune regulation and osteogenic capacity of MSCs in AS are altered by factors such as genetic background, internal environment, infection, and mechanical forces that drive disease development. This review further evaluates the role of MSCs dysfunction in inflammation and pathological bone formation by analyzing the effects of the above-mentioned factors on MSCs function and also looks forward to the prospects of MSCs in treating AS, providing some ideas for an in-depth study of inflammation and ectopic ossification. KEY MESSAGES
Collapse
Affiliation(s)
- Xinzhe Feng
- Department of Joint Bone Disease Surgery, Changhai Hospital, Navy Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Junjie Qiao
- Department of Joint Bone Disease Surgery, Changhai Hospital, Navy Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Weidong Xu
- Department of Joint Bone Disease Surgery, Changhai Hospital, Navy Medical University, 168 Changhai Road, Shanghai 200433, China
| |
Collapse
|
15
|
Shah RJ, Banerjee S, Raychaudhuri S, Raychaudhuri SP. JAK-STAT inhibitors in Immune mediated diseases: An Overview. Indian J Dermatol Venereol Leprol 2023; 89:691-699. [PMID: 37609730 DOI: 10.25259/ijdvl_1152_2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 05/28/2023] [Indexed: 08/24/2023]
Abstract
For any biological response, transmission of extracellular signals to the nucleus is required for DNA transcription and gene expression. In that respect, cytokines/chemokines are well-known inflammatory agents which play a critical role in signalling pathways by activating the Janus kinase-signal transducers and activators of transcription (JAK-STAT) signalling proteins (Janus kinase-signal transducers and activators of transcription) which are a group of intracellular kinase molecules. Cytokines are a category of small proteins (∼5-25 kDa) that play a major role in cell signalling and are major drivers of an autoimmune response. Here we will discuss the role of Janus kinase-signal transducers and activators of transcription kinase cascades in the inflammatory-proliferative cascades of autoimmune disease and about the recent progress in the development of oral synthetic Janus kinase inhibitors (JAKi) and their therapeutic efficacies in dermatologic and systemic autoimmune diseases. Therapeutic efficacy of Janus kinase inhibitors is now well established in the treatment of array of autoimmune and inflammatory disease: spondylarthritis with a special focus on psoriatic arthritis (PsA) and its dermatologic manifestations (psoriasis) and ankylosing spondylitis (AS), atopic dermatitis (AD), alopecia areata (AA), rheumatoid arthritis (RA) and inflammatory bowel disease (IBD). In addition to the first-generation Janus kinase inhibitors several new-generation Janus kinase inhibitors are currently being evaluated. It is expected that these Janus kinase inhibitors likely have higher potency and less adverse effects as compared to their predecessors. Here we have discussed: (1) the functional significance of the Janus kinase-signal transducers and activators of transcription kinase cascades in the inflammatory-proliferative processes of autoimmune diseases and its cellular/molecular mechanisms and (2) progress in the development of oral synthetic Janus kinase inhibitors and their therapeutic efficacies in several systemic and cutaneous autoimmune diseases.
Collapse
Affiliation(s)
- Ruchi Jayesh Shah
- Department of Medicine, School of Medicine, University of California Davis, USA
| | - Sneha Banerjee
- Department of Veterans Affairs, VA Sacramento Medical Center, Northern California Health Care, California, CA, USA
| | - Smriti Raychaudhuri
- Department of Veterans Affairs, VA Sacramento Medical Center, Northern California Health Care, California, CA, USA
| | - Siba P Raychaudhuri
- Department of Medicine, School of Medicine, University of California Davis, USA
| |
Collapse
|
16
|
Mortier C, Gracey E, Coudenys J, Manuello T, Decruy T, Maelegheer M, Stappers F, Gilis E, Gaublomme D, Van Hoorebeke L, Van Welden S, Ambler C, Hegen M, Symanowicz P, Steyn S, Berstein G, Elewaut D, Venken K. RORγt inhibition ameliorates IL-23 driven experimental psoriatic arthritis by predominantly modulating γδ-T cells. Rheumatology (Oxford) 2023; 62:3169-3178. [PMID: 36661300 DOI: 10.1093/rheumatology/kead022] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/19/2022] [Accepted: 01/04/2023] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVE Divergent therapeutic outcomes on different disease domains have been noted with IL-23 and IL-17A-blockade in PsA. Therefore, elucidating the role of RORγt, the master regulator of type 17 immune responses, is of potential therapeutic interest. To this end, RORγt inhibition was assessed in combined skin, joint and gut inflammation in vivo, using a PsA model. METHODS We tested the efficacy of a RORγt antagonist in B10.RIII mice challenged with systemic overexpression of IL-23 by hydrodynamic injection of IL-23 enhanced episomal vector (IL-23 EEV). Clinical outcomes were evaluated by histopathology. Bone density and surface erosions were examined using micro-computed tomography. Cytokine production was measured in serum and by intracellular flow cytometry. Gene expression in PsA-related tissues was analysed by qPCR. RESULTS RORγt-blockade significantly ameliorated psoriasis, peripheral arthritis and colitis development in IL-23 EEV mice (improvement of clinical scores and weight loss respectively by 91.8%, 58.2% and 7.0%, P < 0.001), in line with profound suppression of an enhanced type IL-17 immune signature in PsA-affected tissues. Moreover, inflammation-induced bone loss and bone erosions were reduced (P < 0.05 in calcaneus, P < 0.01 in tibia). Sustained IL-23 overexpression resulted in only mild signs of sacroiliitis. Gamma-delta (γδ)-T cells, the dominant source of T cell-derived IL-17A and IL-22, were expanded during IL-23 overexpression, and together with Th17 cells, clearly countered by RORγt inhibition (P < 0.001). CONCLUSION RORγt-blockade shows therapeutic efficacy in a preclinical PsA model with protection towards extra-musculoskeletal manifestations, reflected by a clear attenuation of type 17 cytokine responses by γδ-T cells and Th17 cells.
Collapse
Affiliation(s)
- Céline Mortier
- Department of Rheumatology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Unit for Molecular Immunology and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Eric Gracey
- Department of Rheumatology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Unit for Molecular Immunology and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Julie Coudenys
- Department of Rheumatology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Unit for Molecular Immunology and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Teddy Manuello
- Department of Rheumatology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Unit for Molecular Immunology and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Tine Decruy
- Department of Rheumatology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Unit for Molecular Immunology and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Margaux Maelegheer
- Department of Rheumatology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Unit for Molecular Immunology and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Flore Stappers
- Department of Rheumatology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Unit for Molecular Immunology and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Elisabeth Gilis
- Department of Rheumatology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Unit for Molecular Immunology and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Djoere Gaublomme
- Department of Rheumatology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Unit for Molecular Immunology and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Luc Van Hoorebeke
- UGCT, Department of Physics and Astronomy, Ghent University, Ghent, Belgium
| | - Sophie Van Welden
- IBD Research Unit, Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Intestinal Barrier Signaling in Disease and Therapy, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | | | - Martin Hegen
- Inflammation and Immunology Research Unit, Pfizer Inc, Cambridge, MA, USA
| | - Peter Symanowicz
- Inflammation and Immunology Research Unit, Pfizer Inc, Cambridge, MA, USA
| | - Stefan Steyn
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc, Cambridge, MA, USA
| | - Gabriel Berstein
- Inflammation and Immunology Research Unit, Pfizer Inc, Cambridge, MA, USA
| | - Dirk Elewaut
- Department of Rheumatology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Unit for Molecular Immunology and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Koen Venken
- Department of Rheumatology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Unit for Molecular Immunology and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| |
Collapse
|
17
|
Cozzi G, Scagnellato L, Lorenzin M, Savarino E, Zingone F, Ometto F, Favero M, Doria A, Vavricka SR, Ramonda R. Spondyloarthritis with inflammatory bowel disease: the latest on biologic and targeted therapies. Nat Rev Rheumatol 2023:10.1038/s41584-023-00984-8. [PMID: 37386288 DOI: 10.1038/s41584-023-00984-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2023] [Indexed: 07/01/2023]
Abstract
Spondyloarthritis (SpA) encompasses a heterogeneous group of chronic inflammatory diseases that can affect both axial and peripheral joints, tendons and entheses. Among the extra-articular manifestations, inflammatory bowel disease (IBD) is associated with considerable morbidity and effects on quality of life. In everyday clinical practice, treatment of these conditions requires a close collaboration between gastroenterologists and rheumatologists to enable early detection of joint and intestinal manifestations during follow-up and to choose the most effective therapeutic regimen, implementing precision medicine for each patient's subtype of SpA and IBD. The biggest issue in this field is the dearth of drugs that are approved for both diseases, as only TNF inhibitors are currently approved for the treatment of full-spectrum SpA-IBD. Janus tyrosine kinase inhibitors are among the most promising drugs for the treatment of peripheral and axial SpA, as well as for intestinal manifestations. Other therapies such as inhibitors of IL-23 and IL-17, phosphodiesterase 4 inhibitor, α4β7 integrin blockers and faecal microbiota transplantation seem to only be able to control some disease domains, or require further studies. Given the growing interest in the development of novel drugs to treat both conditions, it is important to understand the current state of the art and the unmet needs in the management of SpA-IBD.
Collapse
Affiliation(s)
- Giacomo Cozzi
- Rheumatology Unit, Department of Medicine-DIMED, Padova University Hospital, Padova, Italy
| | - Laura Scagnellato
- Rheumatology Unit, Department of Medicine-DIMED, Padova University Hospital, Padova, Italy
| | - Mariagrazia Lorenzin
- Rheumatology Unit, Department of Medicine-DIMED, Padova University Hospital, Padova, Italy
| | - Edoardo Savarino
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University Hospital of Padova, Padova, Italy
| | - Fabiana Zingone
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University Hospital of Padova, Padova, Italy
| | - Francesca Ometto
- Rheumatology Unit, Department of Medicine-DIMED, Padova University Hospital, Padova, Italy
| | - Marta Favero
- Rheumatology Unit, Department of Medicine-DIMED, Padova University Hospital, Padova, Italy
| | - Andrea Doria
- Rheumatology Unit, Department of Medicine-DIMED, Padova University Hospital, Padova, Italy
| | - Stephan R Vavricka
- Department of Gastroenterology and Hepatology, University Hospital Zürich and Center for Gastroenterology and Hepatology, Zürich, Switzerland
| | - Roberta Ramonda
- Rheumatology Unit, Department of Medicine-DIMED, Padova University Hospital, Padova, Italy.
| |
Collapse
|
18
|
Chen B, Jin W. A comprehensive review of stroke-related signaling pathways and treatment in western medicine and traditional Chinese medicine. Front Neurosci 2023; 17:1200061. [PMID: 37351420 PMCID: PMC10282194 DOI: 10.3389/fnins.2023.1200061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/19/2023] [Indexed: 06/24/2023] Open
Abstract
This review provides insight into the complex network of signaling pathways and mechanisms involved in stroke pathophysiology. It summarizes the historical progress of stroke-related signaling pathways, identifying potential interactions between them and emphasizing that stroke is a complex network disease. Of particular interest are the Hippo signaling pathway and ferroptosis signaling pathway, which remain understudied areas of research, and are therefore a focus of the review. The involvement of multiple signaling pathways, including Sonic Hedgehog (SHH), nuclear factor erythroid 2-related factor 2 (Nrf2)/antioxidant response element (ARE), hypoxia-inducible factor-1α (HIF-1α), PI3K/AKT, JAK/STAT, and AMPK in pathophysiological mechanisms such as oxidative stress and apoptosis, highlights the complexity of stroke. The review also delves into the details of traditional Chinese medicine (TCM) therapies such as Rehmanniae and Astragalus, providing an analysis of the recent status of western medicine in the treatment of stroke and the advantages and disadvantages of TCM and western medicine in stroke treatment. The review proposes that since stroke is a network disease, TCM has the potential and advantages of a multi-target and multi-pathway mechanism of action in the treatment of stroke. Therefore, it is suggested that future research should explore more treasures of TCM and develop new therapies from the perspective of stroke as a network disease.
Collapse
Affiliation(s)
- Binhao Chen
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Weifeng Jin
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
19
|
Xue C, Yao Q, Gu X, Shi Q, Yuan X, Chu Q, Bao Z, Lu J, Li L. Evolving cognition of the JAK-STAT signaling pathway: autoimmune disorders and cancer. Signal Transduct Target Ther 2023; 8:204. [PMID: 37208335 DOI: 10.1038/s41392-023-01468-7] [Citation(s) in RCA: 166] [Impact Index Per Article: 83.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 04/22/2023] [Indexed: 05/21/2023] Open
Abstract
The Janus kinase (JAK) signal transducer and activator of transcription (JAK-STAT) pathway is an evolutionarily conserved mechanism of transmembrane signal transduction that enables cells to communicate with the exterior environment. Various cytokines, interferons, growth factors, and other specific molecules activate JAK-STAT signaling to drive a series of physiological and pathological processes, including proliferation, metabolism, immune response, inflammation, and malignancy. Dysregulated JAK-STAT signaling and related genetic mutations are strongly associated with immune activation and cancer progression. Insights into the structures and functions of the JAK-STAT pathway have led to the development and approval of diverse drugs for the clinical treatment of diseases. Currently, drugs have been developed to mainly target the JAK-STAT pathway and are commonly divided into three subtypes: cytokine or receptor antibodies, JAK inhibitors, and STAT inhibitors. And novel agents also continue to be developed and tested in preclinical and clinical studies. The effectiveness and safety of each kind of drug also warrant further scientific trials before put into being clinical applications. Here, we review the current understanding of the fundamental composition and function of the JAK-STAT signaling pathway. We also discuss advancements in the understanding of JAK-STAT-related pathogenic mechanisms; targeted JAK-STAT therapies for various diseases, especially immune disorders, and cancers; newly developed JAK inhibitors; and current challenges and directions in the field.
Collapse
Affiliation(s)
- Chen Xue
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qinfan Yao
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xinyu Gu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qingmiao Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xin Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qingfei Chu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhengyi Bao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
20
|
Bittar M, Mease P. Novel therapies in axial spondyloarthritis. Best Pract Res Clin Rheumatol 2022; 36:101811. [PMID: 36566165 DOI: 10.1016/j.berh.2022.101811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Over the past two decades, advancements in understanding the pathogenesis of axial spondyloarthritis have led to discoveries of new therapeutic targets, particularly the interleukin-17, tumor necrosis factor axis, and Janus kinase-signal transducer and activator of transcription pathway. While many of the available agents have proven to be efficacious and safe for the treatment of axial spondyloarthritis, a remarkable percentage of patients either fail or cannot tolerate these medications. This has prompted researchers to look for new targets that would maximize efficacy and minimize toxicity. In this article, we review novel agents that were recently approved, in trials, and possible future targets or mechanisms. We also discuss their role as it pertains to the prevention of radiographic progression and the management of extra-musculoskeletal manifestations.
Collapse
Affiliation(s)
- Mohamad Bittar
- The University of Tennessee Health Science Center, Division of Connective Tissue Disease (Rheumatology), 956 Court Avenue, Coleman Building, Suite G326, Memphis, TN 38163, USA.
| | - Philip Mease
- Swedish Medical Center/Providence St. Joseph Health and University of Washington School of Medicine, Seattle Rheumatology Associates, 601 Broadway, Suite 600, Seattle, WA 98102, USA.
| |
Collapse
|
21
|
Sen R, Caplan L. Current treatment and molecular targets for axial spondyloarthritis: Evidence from randomized controlled trials. Curr Opin Pharmacol 2022; 67:102307. [PMID: 36335714 DOI: 10.1016/j.coph.2022.102307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 09/20/2022] [Indexed: 11/06/2022]
Abstract
Axial spondyloarthritis (axSpA) is a chronic inflammatory disease that predominantly affects the axial skeleton and is characterized by inflammatory back pain. While much has been published regarding non-steroidal anti-inflammatory drugs and tumor necrosis factor inhibitors, other classes of medications which leverage alternate molecular mechanisms receive less attention. In this review, we summarize a few of the novel targets in axSpA, review the putative mechanism of action of therapies that focus on these targets, and reference the germane recently completed, ongoing, or proposed randomized controlled clinical trials. The agents addressed include inhibitors of interleukin-23, interleukin-17, janus kinases, granulocyte-macrophage colony-stimulating factor, macrophage migration inhibitory factor, antibodies recognizing T cell receptor beta variable 9 gene positive clones, as well as inhibitors of mitogen-activated protein kinase-activated protein kinase-2.
Collapse
Affiliation(s)
- Rouhin Sen
- Rocky Mountain Regional Veterans Affairs Medical Center (VAMC), Denver, CO, USA; University of Colorado School of Medicine, Aurora, CO, USA
| | - Liron Caplan
- Rocky Mountain Regional Veterans Affairs Medical Center (VAMC), Denver, CO, USA; University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
22
|
Rosine N, Rowe H, Koturan S, Yahia‐Cherbal H, Leloup C, Watad A, Berenbaum F, Sellam J, Dougados M, Aimanianda V, Cuthbert R, Bridgewood C, Newton D, Bianchi E, Rogge L, McGonagle D, Miceli‐Richard C. Characterization of Blood Mucosal-Associated Invariant T Cells in Patients With Axial Spondyloarthritis and of Resident Mucosal-Associated Invariant T Cells From the Axial Entheses of Non-Axial Spondyloarthritis Control Patients. Arthritis Rheumatol 2022; 74:1786-1795. [PMID: 35166073 PMCID: PMC9825958 DOI: 10.1002/art.42090] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 12/10/2021] [Accepted: 01/21/2022] [Indexed: 01/11/2023]
Abstract
OBJECTIVE The importance of interleukin-17A (IL-17A) in the pathogenesis of axial spondyloarthritis (SpA) has been demonstrated by the success of IL-17A blockade. However, the nature of the cell populations that produce this important proinflammatory cytokine remains poorly defined. We undertook this study to characterize the major IL-17A-producing blood cell populations in the peripheral blood of patients with axial SpA, with a focus on mucosal-associated invariant T (MAIT) cells, a population known to be capable of producing IL-17. METHODS We evaluated IL-17A production from 5 sorted peripheral blood cell populations, namely, MAIT cells, γδ T cells, CD4+ T cells, CD8+ T cells, and neutrophils, before and after stimulation with phorbol myristate acetate, the calcium ionophore A23187, and β-1,3-glucan. Expression of IL-17A transcripts and protein were determined using nCounter and ultra-sensitive Simoa technology, respectively. MAIT cells from the axial entheses of non-axial SpA control patients (n = 5) were further characterized using flow cytometric immunophenotyping and quantitative polymerase chain reaction, and the production of IL-17 was assessed following stimulation. RESULTS On a per-cell basis, MAIT cells from peripheral blood produced the most IL-17A compared to CD4+ T cells (P < 0.01), CD8+ T cells (P < 0.0001), and γδ T cells (P < 0.0001). IL-17A was not produced by neutrophils. Gene expression analysis also revealed significantly higher expression of IL17A and IL23R in MAIT cells. Stimulation of peripheral blood MAIT cells with anti-CD3/CD28 and IL-7 and/or IL-18 induced strong expression of IL17F. MAIT cells were present in the normal, unaffected entheses of control patients who did not have axial SpA and showed elevated AHR, JAK1, STAT4, and TGFB1 transcript expression with inducible IL-17A protein. IL-18 protein expression was evident in spinal enthesis digests. CONCLUSION Both peripheral blood MAIT cells and resident MAIT cells in normal axial entheses contribute to the production of IL-17 and may play important roles in the pathogenesis of axial SpA.
Collapse
Affiliation(s)
- Nicolas Rosine
- Institut Pasteur, Université de Paris, Immunoregulation Unit, Department of ImmunologyParisFrance
| | - Hannah Rowe
- University of Leeds Institute of Rheumatic and Musculoskeletal MedicineLeedsUK
| | - Surya Koturan
- Institut Pasteur, Université de Paris, Immunoregulation Unit, Department of ImmunologyParisFrance
| | - Hanane Yahia‐Cherbal
- Institut Pasteur, Université de Paris, Immunoregulation Unit, Department of ImmunologyParisFrance
| | - Claire Leloup
- Institut Pasteur, Université de Paris, Immunoregulation Unit, Department of ImmunologyParisFrance
| | - Abdulla Watad
- University of Leeds Institute of Rheumatic and Musculoskeletal MedicineLeedsUK
| | - Francis Berenbaum
- Sorbonne Université, Service de Rhumatologie, Hôpital Saint‐Antoine, AP‐HP, and Centre de Recherche Saint‐Antoine, INSERM UMRS 938ParisFrance
| | - Jeremie Sellam
- Sorbonne Université, Service de Rhumatologie, Hôpital Saint‐Antoine, AP‐HP, and Centre de Recherche Saint‐Antoine, INSERM UMRS 938ParisFrance
| | - Maxime Dougados
- INSERM Unité 1153, Clinical epidemiology and biostatistics, PRES Université Sorbonne Paris Cité, Université de Paris, Service de Rhumatologie, Hôpital Cochin Port Royal, AP‐HP, and Unité Mixte AP‐HP/Institut Pasteur, Institut Pasteur, Immunoregulation UnitParisFrance
| | | | - Richard Cuthbert
- University of Leeds Institute of Rheumatic and Musculoskeletal MedicineLeedsUK
| | - Charlie Bridgewood
- University of Leeds Institute of Rheumatic and Musculoskeletal MedicineLeedsUK
| | - Darren Newton
- University of Leeds Institute of Rheumatic and Musculoskeletal Medicine, Leeds Institute of Medical Research at St James's, and St James's University HospitalLeedsUK
| | - Elisabetta Bianchi
- Institut Pasteur, Université de Paris, Immunoregulation Unit, Department of Immunology, and Unité Mixte AP‐HP/Institut Pasteur, Institut Pasteur, Immunoregulation UnitParisFrance
| | - Lars Rogge
- Institut Pasteur, Université de Paris, Immunoregulation Unit, Department of Immunology, and Unité Mixte AP‐HP/Institut Pasteur, Institut Pasteur, Immunoregulation UnitParisFrance
| | - Dennis McGonagle
- University of Leeds Institute of Rheumatic and Musculoskeletal MedicineLeedsUK
| | - Corinne Miceli‐Richard
- Université de Paris, Service de Rhumatologie, Hôpital Cochin Port Royal, AP‐HP, and Unité Mixte AP‐HP/Institut Pasteur, Institut Pasteur, Immunoregulation UnitParisFrance
| |
Collapse
|
23
|
The JAK-STAT pathway at 30: Much learned, much more to do. Cell 2022; 185:3857-3876. [PMID: 36240739 PMCID: PMC9815833 DOI: 10.1016/j.cell.2022.09.023] [Citation(s) in RCA: 306] [Impact Index Per Article: 102.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/01/2022] [Accepted: 09/14/2022] [Indexed: 11/24/2022]
Abstract
The discovery of the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway arose from investigations of how cells respond to interferons (IFNs), revealing a paradigm in cell signaling conserved from slime molds to mammals. These discoveries revealed mechanisms underlying rapid gene expression mediated by a wide variety of extracellular polypeptides including cytokines, interleukins, and related factors. This knowledge has provided numerous insights into human disease, from immune deficiencies to cancer, and was rapidly translated to new drugs for autoimmune, allergic, and infectious diseases, including COVID-19. Despite these advances, major challenges and opportunities remain.
Collapse
|
24
|
Analysis of N6-Methyladenosine RNA Methylation Regulators in Diagnosis and Distinct Molecular Subtypes of Ankylosing Spondylitis. DISEASE MARKERS 2022; 2022:4942599. [PMID: 36157216 PMCID: PMC9507730 DOI: 10.1155/2022/4942599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 08/17/2022] [Accepted: 08/27/2022] [Indexed: 12/02/2022]
Abstract
The most frequent internal modification in eukaryotic mRNA is N6-methyladenosine (m6A). However, what we know about the m6A regulators in Ankylosing spondylitis (AS) is still limited. In our study, eight distinct m6A regulators were selected utilizing Differentially Expressed Gene (DEG) analysis of the Gene Expression Omnibus GSE73754 dataset for making comparisons between AS (Ankylosing spondylitis) and non-AS patients. The random forest model and the nomogram model were used to screen the eight candidate m6A regulators and evaluate their prediction accuracy for the occurrence of AS. Furthermore, based on the selected m6A regulators, the AS patients were divided into two subgroups, and we applied principal component analysis algorithms to calculate their m6A score and evaluate the m6A patterns. Our findings revealed that patients in cluster A were linked to activated CD4 T cell immunity and activated CD8 T cell immunity. With its major contributions in the area of immunology, our research in m6A patterns may benefit the future diagnosis and treatment strategies of AS.
Collapse
|
25
|
Nguyen CT, Furuya H, Das D, Marusina AI, Merleev AA, Ravindran R, Jalali Z, Khan IH, Maverakis E, Adamopoulos IE. Peripheral γδ T Cells Regulate Neutrophil Expansion and Recruitment in Experimental Psoriatic Arthritis. Arthritis Rheumatol 2022; 74:1524-1534. [PMID: 35320625 PMCID: PMC9427669 DOI: 10.1002/art.42124] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 03/01/2022] [Accepted: 03/17/2022] [Indexed: 11/11/2022]
Abstract
OBJECTIVE This study was undertaken to identify the mechanistic role of γδ T cells in the pathogenesis of experimental psoriatic arthritis (PsA). METHODS In this study, we performed interleukin-23 (IL-23) gene transfer in wild-type (WT) and T cell receptor δ-deficient (TCRδ-/- ) mice and conducted tissue phenotyping in the joint, skin, and nails to characterize the inflammatory infiltrate. We further performed detailed flow cytometry, immunofluorescence staining, RNA sequencing, T cell repertoire analysis, and in vitro T cell polarization assays to identify regulatory mechanisms of γδ T cells. RESULTS We demonstrated that γδ T cells support systemic granulopoiesis, which is critical for murine PsA-like pathology. Briefly, γδ T cell ablation inhibited the expression of neutrophil chemokines CXCL1 and CXCL2 and neutrophil CD11b+Ly6G+ accumulation in the aforementioned PsA-related tissues. Although significantly reduced expression of granulocyte-macrophage colony-stimulating factor (GM-CSF) and IL-17A was detected systemically in TCRδ-/- mice, no GM-CSF+/IL-17A+ γδ T cells were detected locally in the inflamed skin or bone marrow in WT mice. Our data showed that nonresident γδ T cells regulate the expansion of an CD11b+Ly6G+ neutrophil population and their recruitment to joint and skin tissues, where they develop hallmark pathologic features of human PsA. CONCLUSION Our findings do not support the notion that tissue-resident γδ T cells initiate the disease but demonstrate a novel role of γδ T cells in neutrophil regulation that can be exploited therapeutically in PsA patients.
Collapse
Affiliation(s)
- Cuong Thach Nguyen
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis
| | - Hiroki Furuya
- Department of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, USA
| | - Dayasagar Das
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis
| | - Alina I Marusina
- Department of Dermatology, University of California, Davis, Sacramento, CA, USA
| | - Alexander A Merleev
- Department of Dermatology, University of California, Davis, Sacramento, CA, USA
| | - Resmi Ravindran
- Department of Pathology and Laboratory Medicine, University of California at Davis, USA
| | - Zahra Jalali
- Department of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, USA
| | - Imran H. Khan
- Department of Pathology and Laboratory Medicine, University of California at Davis, USA
| | - Emanual Maverakis
- Department of Dermatology, University of California, Davis, Sacramento, CA, USA
| | - Iannis E. Adamopoulos
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis
- Department of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, USA
| |
Collapse
|
26
|
Kenyon M, Maguire S, Rueda Pujol A, O'Shea F, McManus R. The genetic backbone of ankylosing spondylitis: how knowledge of genetic susceptibility informs our understanding and management of disease. Rheumatol Int 2022; 42:2085-2095. [PMID: 35939079 PMCID: PMC9548471 DOI: 10.1007/s00296-022-05174-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 07/20/2022] [Indexed: 11/25/2022]
Abstract
Ankylosing spondylitis (AS) is a seronegative, chronic inflammatory arthritis with high genetic burden. A strong association with HLA-B27 has long been established, but to date its contribution to disease aetiology remains unresolved. Recent insights through genome wide studies reveal an increasing array of immunogenetic risk variants extraneous to the HLA complex in AS cohorts. These genetic traits build a complex profile of disease causality, highlighting several molecular pathways associated with the condition. This and other evidence strongly implicates T-cell-driven pathology, revolving around the T helper 17 cell subset as an important contributor to disease. This prominence of the T helper 17 cell subset has presented the opportunity for therapeutic intervention through inhibition of interleukins 17 and 23 which drive T helper 17 activity. While targeting of interleukin 17 has proven effective, this success has not been replicated with interleukin 23 inhibition in AS patients. Evidence points to significant genetic diversity between AS patients which may, in part, explain the observed refractoriness among a proportion of patients. In this review we discuss the impact of genetics on our understanding of AS and its relationship with closely linked pathologies. We further explore how genetics can be used in the development of therapeutics and as a tool to assist in the diagnosis and management of patients. This evidence indicates that genetic profiling should play a role in the clinician’s choice of therapy as part of a precision medicine strategy towards disease management.
Collapse
Affiliation(s)
- Marcus Kenyon
- Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland.
| | - Sinead Maguire
- Department of Rheumatology, St James' Hospital, Dublin, Ireland
| | - Anna Rueda Pujol
- Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Finbar O'Shea
- Department of Rheumatology, St James' Hospital, Dublin, Ireland
| | - Ross McManus
- Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
27
|
Nielsen OH, Boye TL, Chakravarti D, Gubatan J. Selective tyrosine kinase 2 inhibitors in inflammatory bowel disease. Trends Pharmacol Sci 2022; 43:424-436. [PMID: 35277286 DOI: 10.1016/j.tips.2022.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/09/2022] [Accepted: 02/11/2022] [Indexed: 11/26/2022]
Abstract
Recent significant advances have been made in the treatment of chronic inflammatory diseases with initiation of the era of biologics. However, an unmet medical need still exists for novel targeted therapies. Compared with biologics, Janus kinase inhibitors (JAKis) are a new drug class of orally administered small molecules that have been shown to efficiently modulate complex cytokine-driven inflammation in preclinical models and human studies. Unfortunately, serious adverse effects have been reported with the first introduced pan-JAKi, tofacitinib. Here, we review tyrosine kinase 2 (TYK2) signaling in the pathophysiology of inflammatory bowel disease (IBD), examine mechanisms of action of selective TYK2 inhibitors (TYK2is), and discuss the potential for these inhibitors in efforts to balance benefits and harms.
Collapse
Affiliation(s)
- Ole Haagen Nielsen
- Department of Gastroenterology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark.
| | - Theresa Louise Boye
- Department of Gastroenterology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Deepavali Chakravarti
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John Gubatan
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
28
|
Toussirot E. The Use of Janus Kinase Inhibitors in Axial Spondyloarthritis: Current Insights. Pharmaceuticals (Basel) 2022; 15:ph15030270. [PMID: 35337068 PMCID: PMC8951918 DOI: 10.3390/ph15030270] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 02/17/2022] [Indexed: 01/24/2023] Open
Abstract
Current pharmacological treatments of axial spondyloarthritis (axSpA) are limited to non-steroidal anti-inflammatory drugs (NSAIDs) and biological agents, including TNFα inhibitors and IL-17 inhibitors. Despite the availability of these agents, many patients either fail to respond adequately, lose their initial therapeutic response over time, or develop undesirable side effects, thus highlighting the need for new treatment options. Janus kinase (JAK) and signal transducers and activators of transcription (STAT) are a group of intracellular kinases that play a role in the signaling pathway induced by cytokines and certain growth factors associated with the inflammatory process of axSpA. There are several lines of evidence implicating the JAK–STAT pathway in the pathophysiological process of axSpA, including genetic data, the use of certain JAK in the intracellular signal of specific cytokines involved in axSpA (IL-23, IL-22, and IL-6), and data from experimental models of SpA. This provides a rationale for the assessment of JAK inhibitors (JAKi) in clinical trials with patients with axSpA. In this review, we examine the role of JAK–STAT signaling in the pathogenesis of axSpA and summarize the results from recent clinical trials of JAKi (tofacitinib, upadacitinib, and filgotinib) in patients with axSpA.
Collapse
Affiliation(s)
- Eric Toussirot
- INSERM CIC-1431, Centre d’Investigation Clinique, Pôle Recherche, CHU de Besançon, 25000 Besançon, France;
- Rhumatologie, Pôle PACTE (Pathologies Aiguës Chroniques Transplantation Éducation), CHU de Besançon, 25000 Besançon, France
- Département Universitaire de Thérapeutique, Université de Bourgogne Franche-Comté, 25000 Besançon, France
- INSERM UMR1098 Right “Relations Hôte Greffon Tumeurs, Ingénierie Cellulaire et Génique”, Université de Bourgogne Franche-Comté, 25000 Besançon, France
| |
Collapse
|
29
|
Tofacitinib Blocks Entheseal Lymphocyte Activation and Modulates MSC Adipogenesis, but Does Not Directly Affect Chondro- and Osteogenesis. IMMUNO 2021. [DOI: 10.3390/immuno1040038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Entheseal spinal inflammation and new bone formation with progressive ankylosis may occur in ankylosing spondylitis (AS) and psoriatic arthritis (PsA). This study evaluated whether JAK inhibition with tofacitinib modulated the key disease associated cytokines, TNF and IL-17A, and whether tofacitinib also modulated bone marrow stromal cell-derived mesenchymal stem cell (MSCs) function, including osteogenesis, since post inflammation new bone formation occurs under these conditions. Methods: Conventional entheseal derived αβ CD4+ and CD8+ T-cells were investigated following anti-CD3/CD28 bead stimulation to determine IL-17A and TNF levels in tofacitinib treated (1000 nM) peri-entheseal bone (PEB) and peripheral blood mononuclear cells (PBMC) using ELISA. Bone marrow stromal cell-derived mesenchymal stem cell (MSC) colony forming units (CFU-F) and multi-lineage potential were evaluated using tofacitinib (dosages ranging between 100, 500, 1000 and 10,000 nM). Results: Induced IL-17A and TNF cytokine production from both entheseal CD4+ T-cells and CD8+ T-cells was effectively inhibited by tofacitinib. Tofacitinib treatment did not impact on CFU-F potential or in vitro chondro- and osteogenesis. However, tofacitinib stimulation increased MSC adipogenic potential with greater Oil Red O stained areas. Conclusion: Inducible IL-17A and TNF production by healthy human entheseal CD4+ and CD8+ T-cells was robustly inhibited in vitro by tofacitinib. However, tofacitinib did not impact MSC osteogenesis, but stimulated in vitro MSC adipogenesis, the relevance of which needs further evaluation given that the adipocytes are associated with new bone formation in SpA.
Collapse
|
30
|
Nakamura A, Zeng F, Nakamura S, Reid KT, Gracey E, Lim M, Leng L, Jo S, Park YS, Kusuda M, Machhar R, Boroojeni SF, Wu B, Rossomacha E, Kim TH, Ciccia F, Rockel JS, Kapoor M, Inman RD, Jurisica I, Crome SQ, Bucala R, Haroon N. Macrophage migration inhibitory factor drives pathology in a mouse model of spondyloarthritis and is associated with human disease. Sci Transl Med 2021; 13:eabg1210. [PMID: 34669443 DOI: 10.1126/scitranslmed.abg1210] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Akihiro Nakamura
- Schroeder Arthritis Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada.,Division of Rheumatology, Toronto Western Hospital, University Health Network, Toronto, Ontario M5T 2S8, Canada.,Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Fanxing Zeng
- Schroeder Arthritis Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada
| | - Sayaka Nakamura
- Schroeder Arthritis Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada
| | - Kyle T Reid
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada.,Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, University of Toronto, Toronto, Ontario M5G 2C4, Canada
| | - Eric Gracey
- Unit Molecular Immunology and Inflammation, Inflammation Research Institute, VIB-Ghent University, 9000 Ghent, Belgium.,Department of Rheumatology, Universitair Ziekenhuis Ghent, University of Gent, 9000 Ghent, Belgium
| | - Melissa Lim
- Schroeder Arthritis Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada
| | - Lin Leng
- Section of Rheumatology, Allergy and Immunology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Sungsin Jo
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul 04763, Republic of Korea
| | - Ye-Soo Park
- Department of Orthopaedic Surgery, Guri Hospital, Hanyang University College of Medicine, Guri 11293, Republic of Korea
| | - Masaki Kusuda
- Schroeder Arthritis Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada
| | - Rohan Machhar
- Schroeder Arthritis Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada
| | - Shaghayegh F Boroojeni
- Schroeder Arthritis Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada.,Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Brian Wu
- Schroeder Arthritis Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada
| | - Evgeny Rossomacha
- Schroeder Arthritis Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada
| | - Tae-Hwan Kim
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul 04763, Republic of Korea
| | - Francesco Ciccia
- Department of Precision Medicine, University della Campania L. Vanvitelli, 80131 Naples, Italy
| | - Jason S Rockel
- Schroeder Arthritis Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada
| | - Mohit Kapoor
- Schroeder Arthritis Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada
| | - Robert D Inman
- Schroeder Arthritis Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada.,Division of Rheumatology, Toronto Western Hospital, University Health Network, Toronto, Ontario M5T 2S8, Canada.,Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Igor Jurisica
- Schroeder Arthritis Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada.,Departments of Medical Biophysics and Computer Science, University of Toronto, Toronto, Ontario M5G 1L7, Canada.,Institute of Neuroimmunology, Slovak Academy of Sciences, 85410 Bratislava, Slovakia
| | - Sarah Q Crome
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada.,Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, University of Toronto, Toronto, Ontario M5G 2C4, Canada
| | - Richard Bucala
- Section of Rheumatology, Allergy and Immunology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Nigil Haroon
- Schroeder Arthritis Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada.,Division of Rheumatology, Toronto Western Hospital, University Health Network, Toronto, Ontario M5T 2S8, Canada.,Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
31
|
Ghoreschi K, Augustin M, Baraliakos X, Krönke G, Schneider M, Schreiber S, Schulze-Koops H, Zeißig S, Thaçi D. TYK2‐Inhibition: Potenzial bei der Behandlung chronisch‐entzündlicher Immunerkrankungen. J Dtsch Dermatol Ges 2021; 19:1409-1420. [PMID: 34661350 DOI: 10.1111/ddg.14585_g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 06/08/2021] [Indexed: 12/15/2022]
Affiliation(s)
- Kamran Ghoreschi
- Klinik für Dermatologie, Venerologie und Allergologie, Charité- Universitätsmedizin Berlin
| | - Matthias Augustin
- Institut für Versorgungsforschung in der Dermatologie und bei Pflegeberufen, Universitätsklinikum Hamburg
| | | | - Gerhard Krönke
- Medizinische Klinik 3 (Rheumatologie und Immunologie), Universitätsklinikum Erlangen
| | - Matthias Schneider
- Poliklinik und Funktionsbereich für Rheumatologie, Universitätsklinikum Düsseldorf
| | - Stefan Schreiber
- Institut für Klinische Molekularbiologie, Christian-Albrechts-Universität zu Kiel
| | - Hendrik Schulze-Koops
- Fachbereich für Rheumatologie und Klinische Immunologie, Medizinische Klinik und Poliklinik IV, Universität München
| | - Sebastian Zeißig
- Medizinische Klinik und Poliklinik I - Universitätsklinikum Dresden und Center for Regenerative Therapies Dresden (CRTD)
| | - Diamant Thaçi
- Institut für Entzündungsmedizin, Universitätsklinikum Schleswig- Holstein, Campus Lübeck
| |
Collapse
|
32
|
Simone D, Stingo A, Ciccia F. Genetic and Environmental Determinants of T Helper 17 Pathogenicity in Spondyloarthropathies. Front Genet 2021; 12:703242. [PMID: 34630512 PMCID: PMC8492997 DOI: 10.3389/fgene.2021.703242] [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: 04/30/2021] [Accepted: 08/24/2021] [Indexed: 11/16/2022] Open
Abstract
In Spondyloarthropathies (SpA), a common group of immune-mediated diseases characterised by excessive inflammation of musculo-skeletal structures and extra-articular organs, T helper 17 (Th17) cells are widely considered the main drivers of the disease. Th17 are able to modulate their genes according to the immune environment: upon differentiation, they can adopt either housekeeping, anti-bacterial gene modules or inflammatory, pathogenic functions, and only the latter would mediate immune diseases, such as SpA. Experimental work aimed at characterising Th17 heterogeneity is largely performed on murine cells, for which the in vitro conditions conferring pathogenic potential have been identified and replicated. Interestingly, Th17 recognising different microorganisms are able to acquire specific cytokine signatures. An emerging area of research associates this heterogeneity to the preferential metabolic needs of the cell. In summary, the tissue environment could be determinant for the acquisition of pathogenetic features; this is particularly important at barrier sites, such as the intestine, considered one of the key target organs in SpA, and likely a site of immunological changes that initiate the disease. In this review, we briefly summarise genetic, environmental and metabolic factors that could explain how homeostatic, anti-microbial Th17 could turn into disease-causing cells in Spondyloarthritis.
Collapse
Affiliation(s)
- Davide Simone
- Dipartimento di Medicina di Precisione, Section of RheumatologyUniversità degli Studi della Campania L. Vanvitelli, Naples, Italy
| | - Alessia Stingo
- Dipartimento di Medicina di Precisione, Section of RheumatologyUniversità degli Studi della Campania L. Vanvitelli, Naples, Italy
| | - Francesco Ciccia
- Dipartimento di Medicina di Precisione, Section of RheumatologyUniversità degli Studi della Campania L. Vanvitelli, Naples, Italy
| |
Collapse
|
33
|
Ghoreschi K, Augustin M, Baraliakos X, Krönke G, Schneider M, Schreiber S, Schulze-Koops H, Zeißig S, Thaçi D. TYK2 inhibition and its potential in the treatment of chronic inflammatory immune diseases. J Dtsch Dermatol Ges 2021; 19:1409-1420. [PMID: 34580985 DOI: 10.1111/ddg.14585] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 06/08/2021] [Indexed: 12/12/2022]
Abstract
Immune-mediated chronic inflammatory diseases have emerged as a leading cause of morbidity and mortality in Western countries over the last decades. Although multiple putative factors have been suspected to be causally related to the diseases, their overarching etiology remains unknown. This review article summarizes the current state of scientific knowledge and understanding of the role of non-receptor tyrosine kinases, with a special focus on the Janus kinase TYK2 in autoimmune and immune mediated diseases as well as on the clinical properties of its inhibition. A panel of experts in the field discussed the scientific evidence and molecular rationale for TYK2 inhibition and its clinical application. Reviewing this meeting, we aim at providing an integrated overview of the clinical profile of TYK2 inhibition and its potential in targeted pharmacological therapy of chronic autoimmune and immune-mediated diseases, with a special focus on inflammatory diseases of the skin.
Collapse
Affiliation(s)
- Kamran Ghoreschi
- Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Matthias Augustin
- Institute for Health Services Research in Dermatology and Nursing, University Medical Center Hamburg, Hamburg, Germany
| | - Xenofon Baraliakos
- Center for Rheumatology, Katholische Kliniken Rhein-Ruhr, Herne, Germany
| | - Gerhard Krönke
- Clinic for Inner Medicine 3 (Rheumatology and Immunology), University Hospital Erlangen, Erlangen, Germany
| | - Matthias Schneider
- Polyclinic and Functional Area for Rheumatology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Stefan Schreiber
- Institute of Clinical Molecular Biology, Christian Albrechts University Kiel, Kiel, Germany
| | - Hendrik Schulze-Koops
- Division of Rheumatology and Clinical Immunology, Department of Medicine IV, University of Munich, Munich, Germany
| | - Sebastian Zeißig
- Medical Clinic I - University Hospital Dresden and Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany
| | - Diamant Thaçi
- Institute for Inflammatory Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| |
Collapse
|
34
|
Jo S, Won EJ, Kim MJ, Lee YJ, Jin SH, Park PR, Song HC, Kim J, Choi YD, Kim JY, Shim SC, Choi SH, Park YS, Kim TH, Kim TJ. STAT3 phosphorylation inhibition for treating inflammation and new bone formation in ankylosing spondylitis. Rheumatology (Oxford) 2021; 60:3923-3935. [PMID: 33237331 DOI: 10.1093/rheumatology/keaa846] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 11/19/2020] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE AS is a rheumatic disease characterized by chronic inflammation and bony ankylosis. This study was to evaluate whether a signal transducer and activator of transcription 3 phosphorylation inhibitor (stat3-p Inh) could treat both chronic inflammation and bone formation in AS. METHODS Primary AS osteoprogenitor cells and spinal entheseal cells were examined for osteogenic differentiation. SF mononuclear cells (SFMCs) and lamina propria mononuclear cells (LPMCs) were obtained from AS patients. Inflammatory cytokine-producing cells were analysed using flow cytometry and ELISA. Female SKG mice were treated with stat3-p Inh, IL-17A blocker or vehicle. Inflammation and new bone formation were evaluated using immunohistochemistry, PET and micro-CT. RESULTS In the SKG mouse model, stat3-p Inh significantly suppressed arthritis, enthesitis, spondylitis and ileitis. In experiments culturing SFMCs and LPMCs, the frequencies of IFN-γ-, IL-17A- and TNF-α-producing cells were significantly decreased after stat3-p Inh treatment. When comparing current treatments for AS, stat3-p Inh showed a comparable suppression effect on osteogenesis to Janus kinase inhibitor or IL-17A blocker in AS-osteoprogenitor cells. Stat3-p Inh suppressed differentiation and mineralization of AS-osteoprogenitor cells and entheseal cells toward osteoblasts. Micro-CT analysis of hind paws revealed less new bone formation in stat3-p Inh-treated mice than vehicle-treated mice (P = 0.005). Hind paw and spinal new bone formation were similar between stat3-p Inh- and anti-IL-17A-treated SKG mice (P = 0.874 and P = 0.117, respectively). CONCLUSION Stat-3p inhibition is a promising treatment for both inflammation and new bone formation in AS.
Collapse
Affiliation(s)
- Sungsin Jo
- Hanyang University Institute for Rheumatology Research, Seoul, Republic of Korea
| | - Eun Jeong Won
- Department of Parasitology and Tropical Medicine, Chonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Moon-Ju Kim
- Department of Parasitology and Tropical Medicine, Chonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Yu Jeong Lee
- Department of Parasitology and Tropical Medicine, Chonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - So-Hee Jin
- Department of Rheumatology, Chonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Pu-Reum Park
- Department of Rheumatology, Chonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Ho-Chun Song
- Department of Nuclear Medicine, Chonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Jahae Kim
- Department of Nuclear Medicine, Chonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Yoo-Duk Choi
- Department of Pathology, Chonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Ji-Young Kim
- Division of Rheumatology, Daejeon Rheumatoid & Degenerative Arthritis Center, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Seung Cheol Shim
- Division of Rheumatology, Daejeon Rheumatoid & Degenerative Arthritis Center, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Sung Hoon Choi
- Department of Orthopedic Surgery, Hanyang University Hospital, Seoul, Republic of Korea
| | - Ye-Soo Park
- Department of Orthopedic Surgery, Guri Hospital, Hanyang University College of Medicine, Guri, Republic of Korea
| | - Tae-Hwan Kim
- Hanyang University Institute for Rheumatology Research, Seoul, Republic of Korea.,Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, Republic of Korea
| | - Tae-Jong Kim
- Department of Rheumatology, Chonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| |
Collapse
|
35
|
Baglaenko Y, Macfarlane D, Marson A, Nigrovic PA, Raychaudhuri S. Genome editing to define the function of risk loci and variants in rheumatic disease. Nat Rev Rheumatol 2021; 17:462-474. [PMID: 34188205 PMCID: PMC10782829 DOI: 10.1038/s41584-021-00637-8] [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] [Accepted: 05/20/2021] [Indexed: 02/06/2023]
Abstract
Discoveries in human genetic studies have revolutionized our understanding of complex rheumatic and autoimmune diseases, including the identification of hundreds of genetic loci and single nucleotide polymorphisms that potentially predispose individuals to disease. However, in most cases, the exact disease-causing variants and their mechanisms of action remain unresolved. Functional follow-up of these findings is most challenging for genomic variants that are in non-coding genomic regions, where the large majority of common disease-associated variants are located, and/or that probably affect disease progression via cell type-specific gene regulation. To deliver on the therapeutic promise of human genetic studies, defining the mechanisms of action of these alleles is essential. Genome editing technology, such as CRISPR-Cas, has created a vast toolbox for targeted genetic and epigenetic modifications that presents unprecedented opportunities to decipher disease-causing loci, genes and variants in autoimmunity. In this Review, we discuss the past 5-10 years of progress in resolving the mechanisms underlying rheumatic disease-associated alleles, with an emphasis on how genomic editing techniques can enable targeted dissection and mechanistic studies of causal autoimmune risk variants.
Collapse
Affiliation(s)
- Yuriy Baglaenko
- Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Dana Macfarlane
- Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Alexander Marson
- Gladstone Institutes, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Department of Microbiology and Immunology, University of California, San Francisco, CA, USA
- Innovative Genomics Institute, University of California, Berkeley, CA, USA
- Rosalind Russell/Ephraim P. Engleman Rheumatology Research Center, University of California, San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Peter A Nigrovic
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Division of Immunology, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Soumya Raychaudhuri
- Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA.
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
- Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK.
| |
Collapse
|
36
|
Hromadová D, Elewaut D, Inman RD, Strobl B, Gracey E. From Science to Success? Targeting Tyrosine Kinase 2 in Spondyloarthritis and Related Chronic Inflammatory Diseases. Front Genet 2021; 12:685280. [PMID: 34290741 PMCID: PMC8287328 DOI: 10.3389/fgene.2021.685280] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/02/2021] [Indexed: 12/16/2022] Open
Abstract
Spondyloarthritis (SpA) is a family of inflammatory arthritic diseases, which includes the prototypes of psoriatic arthritis and ankylosing spondylitis. SpA is commonly associated with systemic inflammatory diseases, such as psoriasis and inflammatory bowel disease. Immunological studies, murine models and the genetics of SpA all indicate a pathogenic role for the IL-23/IL-17 axis. Therapeutics targeting the IL-23/IL-17 pathway are successful at providing symptomatic relief, but may not provide complete protection against progression of arthritis. Thus there is still tremendous interest in the discovery of novel therapeutic targets for SpA. Tyrosine kinase 2 (TYK2) is a member of the Janus kinases, which mediate intracellular signaling of cytokines via signal transducer and activator of transcription (STAT) activation. TYK2 plays a crucial role in mediating IL-23 receptor signaling and STAT3 activation. A plethora of natural mutations in and around TYK2 have provided a wealth of data to associate this kinase with autoimmune/autoinflammatory diseases in humans. Induced and natural mutations in murine Tyk2 largely support human data; however, key inter-species differences exist, which means extrapolation of data from murine models to humans needs to be done with caution. Despite these reservations, novel selective TYK2 inhibitors are now proving successful in advanced clinical trials of inflammatory diseases. In this review, we will discuss TYK2 from basic biology to therapeutic targeting, with an emphasis on studies in SpA. Seminal studies uncovering the basic science of TYK2 have provided sound foundations for targeting it in SpA and related inflammatory diseases. TYK2 inhibitors may well be the next blockbuster therapeutic for SpA.
Collapse
Affiliation(s)
- Dominika Hromadová
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Dirk Elewaut
- Molecular Immunology and Inflammation Unit, VIB Centre for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Rheumatology, Ghent University Hospital, Ghent, Belgium
| | - Robert D. Inman
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada
- Departments of Medicine and Immunology, University of Toronto, Toronto, ON, Canada
| | - Birgit Strobl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Eric Gracey
- Molecular Immunology and Inflammation Unit, VIB Centre for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Rheumatology, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
37
|
Raychaudhuri S, Cheema KS, Raychaudhuri SK, Raychaudhuri SP. Janus kinase-signal transducers and activators of transcription cell signaling in Spondyloarthritis: rationale and evidence for JAK inhibition. Curr Opin Rheumatol 2021; 33:348-355. [PMID: 34014847 DOI: 10.1097/bor.0000000000000810] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
PURPOSE OF REVIEW The Janus kinase-signal transducers and activators of transcription (JAK-STAT) signaling proteins represent a group of intracellular kinase molecules that play a central role in the signaling pathways induced by cytokines, chemokines, and certain growth factors associated with systemic and local inflammation of autoimmune diseases including in Spondyloarthritis (SpA). Here, we will discuss (i) the functional significance of the JAK-STAT kinase cascades in the inflammatory-proliferative processes of SpA and its cellular/molecular mechanisms (ii) progress in the development of oral synthetic JAK inhibitors (JAKi) and their therapeutic efficacies in SpA. RECENT FINDINGS Development JAKi is a fast-moving field in the medical science. Several new-generation JAKi are being identified for psoriatic arthritis and ankylosing spondylitis. It is expected these JAKi likely to have higher potency and less adverse effects. SUMMARY Here, we are providing an updated review on the significance of JAK-STAT signaling proteins in SpA with an emphasis on new-generation of JAK-STAT inhibitors for the treatment of SpA.
Collapse
Affiliation(s)
| | | | - Smriti K Raychaudhuri
- VA Sacramento Medical Center, Department of Veterans Affairs, Northern California Healthcare System, Mather
- School of Medicine, University of California, Davis, California, USA
| | - Siba P Raychaudhuri
- VA Sacramento Medical Center, Department of Veterans Affairs, Northern California Healthcare System, Mather
- School of Medicine, University of California, Davis, California, USA
| |
Collapse
|
38
|
Mandour M, Chen S, van de Sande MGH. The Role of the IL-23/IL-17 Axis in Disease Initiation in Spondyloarthritis: Lessons Learned From Animal Models. Front Immunol 2021; 12:618581. [PMID: 34267743 PMCID: PMC8276000 DOI: 10.3389/fimmu.2021.618581] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 05/31/2021] [Indexed: 12/14/2022] Open
Abstract
Spondyloarthritis (SpA) is a spectrum of chronic inflammatory joint diseases that frequently presents with inflammation of the axial skeleton, peripheral joints, entheses, skin, and gut. Understanding SpA pathogenesis has been proven challenging due to the limited availability of human target tissues. In recent years, the interleukin (IL)-23/IL-17 pathway has been implicated in the pathogenesis of SpA, in addition to the Tumor Necrosis Factor Alpha (TNF-α) cytokine. The underlying molecular mechanisms by which the IL-23/IL-17 pathway triggers disease initiation, both in the joints as well as at extra-musculoskeletal sites, are not precisely known. Animal models that resemble pathological features of human SpA have provided possibilities for in-depth molecular analyses of target tissues during various phases of the disease, including the pre-clinical initiation phase of the disease before arthritis and spondylitis are clinically present. Herein, we summarize recent insights gained in SpA animal models on the role of the IL-23/IL-17 pathway in immune activation across affected sites in SpA, which include the joint, entheses, gut and skin. We discuss how local activation of the IL-23/IL-17 axis may contribute to the development of tissue inflammation and the onset of clinically manifest SpA. The overall aim is to provide the reader with an overview of how the IL-23/IL-17 axis could contribute to the onset of SpA pathogenesis. We discuss how insights from animal studies into the initiation phase of disease could instruct validation studies in at-risk individuals and thereby provide a perspective for potential future preventive treatment.
Collapse
Affiliation(s)
- Mohamed Mandour
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Immunology, Infection and Immunity Institute, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Sijia Chen
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Immunology, Infection and Immunity Institute, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam, Netherlands
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Marleen G. H. van de Sande
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Immunology, Infection and Immunity Institute, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
39
|
Ankylosing spondylitis: an autoimmune or autoinflammatory disease? Nat Rev Rheumatol 2021; 17:387-404. [PMID: 34113018 DOI: 10.1038/s41584-021-00625-y] [Citation(s) in RCA: 173] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2021] [Indexed: 12/20/2022]
Abstract
Ankylosing spondylitis (AS) is a chronic inflammatory disorder of unknown aetiology. Unlike other systemic autoimmune diseases, in AS, the innate immune system has a dominant role characterized by aberrant activity of innate and innate-like immune cells, including γδ T cells, group 3 innate lymphoid cells, neutrophils, mucosal-associated invariant T cells and mast cells, at sites predisposed to the disease. The intestine is involved in disease manifestations, as it is at the forefront of the interaction between the mucosal-associated immune cells and the intestinal microbiota. Similarly, biomechanical factors, such as entheseal micro-trauma, might also be involved in the pathogenesis of the articular manifestation of AS, and sentinel immune cells located in the entheses could provide links between local damage, genetic predisposition and the development of chronic inflammation. Although these elements might support the autoinflammatory nature of AS, studies demonstrating the presence of autoantibodies (such as anti-CD74, anti-sclerostin and anti-noggin antibodies) and evidence of activation and clonal expansion of T cell populations support an autoimmune component to the disease. This Review presents the evidence for autoinflammation and the evidence for autoimmunity in AS and, by discussing the pathophysiological factors associated with each, aims to reconcile the two hypotheses.
Collapse
|
40
|
Ceribelli A, Motta F, Vecellio M, Isailovic N, Ciccia F, Selmi C. Clinical Trials Supporting the Role of the IL-17/IL-23 Axis in Axial Spondyloarthritis. Front Immunol 2021; 12:622770. [PMID: 34149686 PMCID: PMC8206811 DOI: 10.3389/fimmu.2021.622770] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 05/14/2021] [Indexed: 12/21/2022] Open
Abstract
The term spondyloarthritis (SpA) encompasses a heterogeneous group of inflammatory musculoskeletal diseases with several common genetic background and clinical features, including the possible involvement of the axial skeleton with peripheral mono- or oligo- arthritis and frequently coexisting skin, eye and intestinal manifestations. When the sacroiliac joints or other parts of the spine or thoracic wall are predominantly affected at magnetic resonance or X-ray imaging with inflammatory back pain, the disease is classified as axial SpA and the therapeutic choices are significantly different compared to cases of peripheral arthritis. Moving from the narrow effectiveness and safety profiles of non-steroidal anti-inflammatory drugs, there has been a significant research effort aimed at identifying new treatments based on our better understanding of the pathogenesis of SpA. Indeed, in parallel with the solid data demonstrating that IL-17 and IL-23 are key cytokines in the development of enthesitis and spondylitis, monoclonal antibodies interfering with this pathway have been developed for the treatment of axial SpA. Furthermore, the IL-17/IL-23 axis is key to extra-articular manifestations such as inflammatory bowel disease, uveitis, and psoriasis which are frequent comorbidities of SpA. Currently available drugs act through these mechanisms recognizing IL-23 and targeting IL-17, such as secukinumab and ixekizumab. These therapeutic approaches are now envisioned in the international treatment recommendations for psoriatic arthritis with an axial phenotype as well as for ankylosing spondylitis (AS). We will provide herein a concise comprehensive overview of the clinical evidence supporting the use of these and other drugs acting on IL-23 and IL-17 in axial SpA.
Collapse
Affiliation(s)
- Angela Ceribelli
- Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center - IRCCS, Rozzano (Mi), Italy
- Department of Biomedical Sciences, Humanitas University, Rozzano (Mi), Italy
| | - Francesca Motta
- Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center - IRCCS, Rozzano (Mi), Italy
- Department of Biomedical Sciences, Humanitas University, Rozzano (Mi), Italy
| | - Matteo Vecellio
- Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center - IRCCS, Rozzano (Mi), Italy
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Natasa Isailovic
- Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center - IRCCS, Rozzano (Mi), Italy
| | - Francesco Ciccia
- Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Carlo Selmi
- Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center - IRCCS, Rozzano (Mi), Italy
- Department of Biomedical Sciences, Humanitas University, Rozzano (Mi), Italy
| |
Collapse
|
41
|
Łukasik Z, Gracey E, Venken K, Ritchlin C, Elewaut D. Crossing the boundaries: IL-23 and its role in linking inflammation of the skin, gut and joints. Rheumatology (Oxford) 2021; 60:iv16-iv27. [PMID: 33961030 PMCID: PMC8527243 DOI: 10.1093/rheumatology/keab385] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/17/2021] [Indexed: 11/17/2022] Open
Abstract
Several lines of evidence point towards the central role of IL-23 as a crucial inflammatory mediator in the pathogenesis of SpA—a group of inflammatory arthritic diseases whose symptoms span the skin, gastrointestinal tract and joints. While therapeutic blockade of IL-23 proved successful in the treatment of IBD, psoriatic skin disease and peripheral SpA, it failed in patients suffering from SpA with predominantly axial involvement. Here we review state-of-the-art discoveries on IL-23 signalling pathways across target tissues involved in SpA. We discuss the discrepancies in resident IL-23–responding cells and their downstream activities across skin, gut and joint that shape the unique immunological landscape of SpA.
Collapse
Affiliation(s)
- Zuzanna Łukasik
- Department of Internal Medicine and Pediatrics, UZ Ghent, Ghent University, Ghent, Belgium.,VIB Center for Inflammation Research, Ghent University, Belgium
| | - Eric Gracey
- Department of Internal Medicine and Pediatrics, UZ Ghent, Ghent University, Ghent, Belgium.,VIB Center for Inflammation Research, Ghent University, Belgium
| | - Koen Venken
- Department of Internal Medicine and Pediatrics, UZ Ghent, Ghent University, Ghent, Belgium.,VIB Center for Inflammation Research, Ghent University, Belgium
| | - Christopher Ritchlin
- Department of Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Dirk Elewaut
- Department of Internal Medicine and Pediatrics, UZ Ghent, Ghent University, Ghent, Belgium.,VIB Center for Inflammation Research, Ghent University, Belgium.,Ghent Gut Inflammation Group, Ghent University, Ghent, Belgium
| |
Collapse
|
42
|
Nakamura A, Haroon N. Recent Updates in the Immunopathology of Type 3 Immunity-Mediated Enthesitis. Curr Rheumatol Rep 2021; 23:31. [PMID: 33893896 DOI: 10.1007/s11926-021-00995-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2021] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW Enthesitis is a cardinal feature of spondyloarthritis (SpA). Despite increasing available treatments, challenges remain in adequately controlling inflammation and subsequent new bone formation (NBF) in entheses; thus, a better understanding of the immunopathogenesis is warranted. RECENT FINDINGS Increasing evidence has identified immune cells playing key roles in enthesitis such as γδ T cells and group 3 innate lymphoid cells (ILC3), possibly with site-specific regulatory systems. The presence of T cells producing interleukin (IL)-17 independent of IL-23 in human spinal entheses was recently reported, which may corroborate the discrepancy between recent clinical trials and pre-clinical studies. In addition, the contribution of myeloid cells has also been focused in both human and pre-clinical SpA models. Moreover, not only the IL-23/IL-17 signaling, but other key type 3 immunity mediators, such as IL-22 and granulocyte-macrophage colony-stimulating factor (GM-CSF), have been reported as pivotal cytokines in inflammation and NBF of entheses. Immune cells demonstrating distinct features orchestrate entheses, leading to the complex landscape of enthesitis. However, recent advances in understanding the immunopathogenesis may provide new therapeutic targets and future research directions.
Collapse
Affiliation(s)
- Akihiro Nakamura
- Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada.,Spondylitis Program, University Health Network, Toronto, Ontario, Canada.,Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Division of Rheumatology, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Nigil Haroon
- Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada. .,Spondylitis Program, University Health Network, Toronto, Ontario, Canada. .,Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada. .,Division of Rheumatology, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada. .,Institute of Medical Science, Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
43
|
Inman RD. Axial Spondyloarthritis: Current Advances, Future Challenges. JOURNAL OF RHEUMATIC DISEASES 2021; 28:55-59. [PMID: 37476012 PMCID: PMC10324891 DOI: 10.4078/jrd.2021.28.2.55] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/17/2021] [Accepted: 03/10/2021] [Indexed: 07/22/2023]
Abstract
Axial spondyloarthritis (axSpA) is a chronic inflammatory joint disease with a predilection for the spine. It affects young adults and has the potential to have a major impact on quality of life, not only because of the chronic pain and fatigue, but also because of the potential for marked disability related to spinal ankylosis. Early detection of axSpA remains a major challenge, for which there is a heightened sense of urgency since it has been shown that earlier intervention with biologics can alter the progression of radiographic change in the spine. Advances in the genetics of axSpA have highlighted a number of candidate genes conferring susceptibility to the disease, but there is evidence of environmental factors playing a role as well. Recently studies in both clinical and experimental axSpA have implicated alterations in the gut microbiome as playing a key role, and the immunology of the gut-joint axis is becoming better understood. The unmet needs which are shaping the research agenda include improvement in early case identification, sensitive and specific biomarkers which could accurately reflect disease activity and severity, improved understanding of the common pathways of inflammation in the skin, eye and gut in axSpA, and novel therapeutic targets which could have curative potential.
Collapse
Affiliation(s)
- Robert D Inman
- Schroeder Arthritis Institute, University Health Network and University of Toronto, Toronto, ON, Canada
| |
Collapse
|
44
|
McGonagle D, Watad A, Sharif K, Bridgewood C. Why Inhibition of IL-23 Lacked Efficacy in Ankylosing Spondylitis. Front Immunol 2021; 12:614255. [PMID: 33815371 PMCID: PMC8017223 DOI: 10.3389/fimmu.2021.614255] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 01/18/2021] [Indexed: 12/24/2022] Open
Abstract
The term spondyloarthritis pertains to both axial and peripheral arthritis including ankylosing spondylitis (AS) and psoriatic arthritis (PsA), which is strongly linked to psoriasis and also the arthritis associated with inflammatory bowel disease. The argument supporting the role for IL-23 across the spectrum of SpA comes from 4 sources. First, genome wide associated studies (GWAS) have shown that all the aforementioned disorders exhibit IL-23R pathway SNPs, whereas HLA-B27 is not linked to all of these diseases-hence the IL-23 pathway represents the common genetic denominator. Secondly, experimental animal models have demonstrated a pivotal role for the IL-23/IL-17 axis in SpA related arthropathy that initially manifests as enthesitis, but also synovitis and axial inflammation and also associated aortic root and cutaneous inflammation. Thirdly, the emergent immunology of the human enthesis also supports the presence of IL-23 producing myeloid cells, not just at the enthesis but in other SpA associated sites including skin and gut. Finally, drugs that target the IL-23 pathway show excellent efficacy for skin disease, efficacy for IBD and also in peripheral arthropathy associated with SpA. The apparent failure of IL-23 blockade in the AS which is effectively a spinal polyenthesitis but evidence for efficacy of IL-23 inhibition for peripheral enthesitis in PsA and preliminary suggestions for benefit in axial PsA, raises many questions. Key amongst these is whether spinal inflammation may exhibit entheseal IL-17A production independent of IL-23 but peripheral enthesitis is largely dependent on IL-23 driven IL-17 production. Furthermore, IL-23 blocking strategies in animal models may prevent experimental SpA evolution but not prevent established disease, perhaps pointing towards a role for IL-23 in innate immune disease initiation whereas persistent disease is dependent on memory T-cell responses that drive IL-17A production independently of IL-23, but this needs further study. Furthermore, IL-12/23 posology in inflammatory bowel disease is substantially higher than that used in AS trials which merits consideration. Therefore, the IL-23 pathway is centrally involved in the SpA concept but the nuances and intricacies in axial inflammation that suggest non-response to IL-23 antagonism await formal definition. The absence of comparative immunology between the different skeletal sites renders explanations purely hypothetical at this juncture.
Collapse
Affiliation(s)
- Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
- National Institute for Health Research (NIHR), Leeds Biomedical Research Centre (BRC), Leeds Teaching Hospitals, Leeds, United Kingdom
| | - Abdulla Watad
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
- Department of Medicine ‘B’, Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Kassem Sharif
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
- Department of Medicine ‘B’, Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Charlie Bridgewood
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
45
|
Mucosal Epithelial Jak Kinases in Health and Diseases. Mediators Inflamm 2021; 2021:6618924. [PMID: 33814980 PMCID: PMC7990561 DOI: 10.1155/2021/6618924] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/02/2021] [Accepted: 03/06/2021] [Indexed: 12/22/2022] Open
Abstract
Janus kinases (Jaks) are a family of nonreceptor tyrosine kinase that include four different members, viz., Jak1, Jak2, Jak3, and Tyk2. Jaks play critical roles in immune cells functions; however, recent studies suggest they also play essential roles in nonimmune cell physiology. This review highlights the significance of epithelial Jaks in understanding the molecular basis of some of the diseases through regulation of epithelial-mesenchymal transition, cell survival, cell growth, development, and differentiation. Growth factors and cytokines produced by the cells of hematopoietic origin use Jak kinases for signal transduction in both immune and nonimmune cells. Among Jaks, Jak3 is widely expressed in both immune cells and in intestinal epithelial cells (IECs) of both humans and mice. Mutations that abrogate Jak3 functions cause an autosomal severe combined immunodeficiency disease (SCID) while activating Jak3 mutations lead to the development of hematologic and epithelial cancers. A selective Jak3 inhibitor CP-690550 (Xeljanz) approved by the FDA for certain chronic inflammatory conditions demonstrates immunosuppressive activity in rheumatoid arthritis, psoriasis, and organ transplant rejection. Here, we also focus on the consequences of Jak3-directed drugs on adverse effects in light of recent discoveries in mucosal epithelial functions of Jak3 with some information on other Jaks. Lastly, we brief on structural implications of Jak3 domains beyond the immune cells. As information about the roles of Jak3 in gastrointestinal functions and associated diseases are only just emerging, in the review, we summarize its implications in gastrointestinal wound repair, inflammatory bowel disease, obesity-associated metabolic syndrome, and epithelial cancers. Lastly, we shed lights on identifying potential novel targets in developing therapeutic interventions of diseases associated with dysfunctional IEC.
Collapse
|
46
|
Lee YJ, Kim MJ, Jo S, Jin SH, Park PR, Park K, Song HC, Kim J, Kim JY, Shim SC, Kim TH, Hong SJ, Kang H, Kim TJ, Won EJ. Clonorchis sinensis-Derived Protein Attenuates Inflammation and New Bone Formation in Ankylosing Spondylitis. Front Immunol 2021; 12:615369. [PMID: 33717104 PMCID: PMC7947613 DOI: 10.3389/fimmu.2021.615369] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 01/18/2021] [Indexed: 12/17/2022] Open
Abstract
Helminth infections and their components have been shown to have the potential to modulate and attenuate immune responses. The objective of this study was to evaluate the potential protective effects of Clonorchis sinensis-derived protein (CSp) on ankylosing spondylitis (AS). Cytotoxicity of CSp at different doses was assessed by MTS and flow cytometry before performing experiments. Peripheral blood mononuclear cells (PBMCs) and synovial fluid mononuclear cells (SFMCs) were obtained from AS patients. Inflammatory cytokine-producing cells were analyzed using flow cytometry. The levels of INF- γ , IL-17A, TNF-α, and IL-6 were measured by enzyme-linked immunosorbent assay (ELISA). SKG mice were treated with CSp or vehicles. Inflammation and new bone formation were evaluated using immunohistochemistry, positron emission tomography (PET), and micro-computed tomography (CT). Treatment with CSp resulted in no reduced cell viability of PBMCs or SFMCs until 24 h. In experiments culturing PBMCs and SFMCs, the frequencies of IFN- γ and IL-17A producing cells were significantly reduced after CSp treatment. In the SKG mouse model, CSp treatment significantly suppressed arthritis, enthesitis, and enteritis. Micro-CT analysis of hind paw revealed reduced new bone formation in CSp-treated mice than in vehicle-treated mice. We provide the first evidence demonstrating that CSp can ameliorate clinical signs and cytokine derangements in AS. In addition, such CSp treatment could reduce the new bone formation of AS.
Collapse
Affiliation(s)
- Yu Jeong Lee
- Department of Parasitology and Tropical Medicine, Chonnam National University Medical School, Gwangju, South Korea
| | - Moon-Ju Kim
- Department of Parasitology and Tropical Medicine, Chonnam National University Medical School, Gwangju, South Korea
| | - Sungsin Jo
- Department of Rheumatology, Hanyang University Institute for Rheumatology Research, Seoul, South Korea
| | - So-Hee Jin
- Department of Rheumatology, Chonnam National University Medical School and Hospital, Gwangju, South Korea
| | - Pu-Reum Park
- Department of Rheumatology, Chonnam National University Medical School and Hospital, Gwangju, South Korea
| | - Kijeong Park
- Department of Rheumatology, Chonnam National University Medical School and Hospital, Gwangju, South Korea
| | - Ho-Chun Song
- Department of Nuclear Medicine, Chonnam National University Medical School and Hospital, Gwangju, South Korea
| | - Jahae Kim
- Department of Nuclear Medicine, Chonnam National University Medical School and Hospital, Gwangju, South Korea
| | - Ji-Young Kim
- Division of Rheumatology, Daejeon Rheumatoid & Degenerative Arthritis Center, Chungnam National University Hospital, Daejeon, South Korea
| | - Seung Cheol Shim
- Division of Rheumatology, Daejeon Rheumatoid & Degenerative Arthritis Center, Chungnam National University Hospital, Daejeon, South Korea
| | - Tae-Hwan Kim
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, South Korea
| | - Sung-Jong Hong
- Department of Medical Environmental Biology, Chung-Ang University College of Medicine, Seoul, South Korea
| | - Hyundeok Kang
- Department of Biomedical Systems Informatics, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Tae-Jong Kim
- Department of Rheumatology, Chonnam National University Medical School and Hospital, Gwangju, South Korea
| | - Eun Jeong Won
- Department of Parasitology and Tropical Medicine, Chonnam National University Medical School, Gwangju, South Korea
| |
Collapse
|
47
|
Pyroptosis by caspase-11 inflammasome-Gasdermin D pathway in autoimmune diseases. Pharmacol Res 2021; 165:105408. [PMID: 33412278 DOI: 10.1016/j.phrs.2020.105408] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/28/2020] [Accepted: 12/28/2020] [Indexed: 02/08/2023]
Abstract
Inflammasomes are a group of supramolecular complexes primarily comprise a sensor, adaptor protein and an effector. Among them, canonical inflammasomes are assembled by one specific pattern recognition receptor, the adaptor protein apoptosis-associated speck-like protein containing a CARD and procaspase-1. Murine caspase-11 and its human ortholog caspase-4/5 are identified as cytosolic sensors which directly responds to LPS. Once gaining access to cytosol, LPS further trigger inflammasome activation in noncanonical way. Downstream pore-forming Gasdermin D is a pyroptosis executioner. Emerging evidence announced in recent years demonstrate the vital role played by caspase-11 non-canonical inflammasome in a range of autoimmune diseases. Pharmacological ablation of caspase-11 and its related effector results in potent therapeutic effects. Though recent advances have highlighted the potential of caspase-11 as a drug target, the understanding of caspase-11 molecular activation and regulation mechanism remains to be limited and thus hampered the discovery and progression of novel inhibitors. Here in this timeline review, we explored how caspase-11 get involved in the pathogenesis of autoimmune diseases, we also collected the reported small-molecular caspase-11 inhibitors. Moreover, the clinical implications and therapeutic potential of caspase-11 inhibitors are discussed. Targeting non-canonical inflammasomes is a promising strategy for autoimmune diseases treatment, while information about the toxicity and physiological disposition of the promising caspase-11 inhibitors need to be supplemented before they can be translated from bench to bedside.
Collapse
|
48
|
Hammitzsch A, Lorenz G, Moog P. Impact of Janus Kinase Inhibition on the Treatment of Axial Spondyloarthropathies. Front Immunol 2020; 11:591176. [PMID: 33193430 PMCID: PMC7609840 DOI: 10.3389/fimmu.2020.591176] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/07/2020] [Indexed: 12/30/2022] Open
Abstract
Many immune cells and effector molecules (e.g. cytokines, Interferons, growth factors) utilize different combinations of Janus kinase (JAK) and signal transducer and activator of transcription (STAT) molecules to transduce signals from the cell surface to the nucleus, where they regulate transcription. This pathway is basically involved in almost all inflammatory diseases and also in the interleukin (IL)-23/IL-17 cascade, which is an essential part of the pathogenesis of spondyloarthropathies (SpA). Upon evidence from in vitro and in vivo experiments indicating disease-modifying effects of JAK inhibition in inflammatory joint disease, numerous inhibitors of the JAK/STAT pathway (= JAKinibs) with different selectivity against the four members of the JAK family [JAK1, JAK2, JAK3, and tyrosine kinase 2 (TYK2)] were developed. Trials in rheumatoid arthritis were successful with respect to efficacy and safety, and currently, three JAKinibs are approved for the treatment of rheumatoid arthritis in the European Union. Although new treatment options (anti-IL-23, anti-IL-17, and phosphodiesterase 4 inhibitors) have become available for spondyloarthritis and especially psoriatic arthritis (PsA) within the last years, most of them are biologics and do not address all disease manifestations equally. Therefore, multiple trials were initiated to evaluate JAKinibs in PsA and axial spondyloarthritis (axSpA). A trial of Tofacitinib (OPAL) was successful in PsA and has led to the inclusion of JAKinibs into the treatment algorithm. Currently many trials with JAKinibs are ongoing for PsA and axSpA, with one phase III trial of upadacitinib (selective JAK1 inhibitor) showing good therapeutic response in active radiographic axSpA.
Collapse
Affiliation(s)
- Ariane Hammitzsch
- Section of Rheumatology, Department of Nephrology, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Georg Lorenz
- Section of Rheumatology, Department of Nephrology, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Philipp Moog
- Section of Rheumatology, Department of Nephrology, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| |
Collapse
|
49
|
IL-12 and IL-23-Close Relatives with Structural Homologies but Distinct Immunological Functions. Cells 2020; 9:cells9102184. [PMID: 32998371 PMCID: PMC7600943 DOI: 10.3390/cells9102184] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 09/24/2020] [Accepted: 09/27/2020] [Indexed: 12/21/2022] Open
Abstract
Cytokines of the IL-12 family show structural similarities but have distinct functions in the immune system. Prominent members of this cytokine family are the pro-inflammatory cytokines IL-12 and IL-23. These two cytokines share cytokine subunits and receptor chains but have different functions in autoimmune diseases, cancer and infections. Accordingly, structural knowledge about receptor complex formation is essential for the development of new therapeutic strategies preventing and/or inhibiting cytokine:receptor interaction. In addition, intracellular signaling cascades can be targeted to inhibit cytokine-mediated effects. Single nucleotide polymorphisms can lead to alteration in the amino acid sequence and thereby influencing protein functions or protein–protein interactions. To understand the biology of IL-12 and IL-23 and to establish efficient targeting strategies structural knowledge about cytokines and respective receptors is crucial. A highly efficient therapy might be a combination of different drugs targeting extracellular cytokine:receptor assembly and intracellular signaling pathways.
Collapse
|
50
|
TYK2 licenses non-canonical inflammasome activation during endotoxemia. Cell Death Differ 2020; 28:748-763. [PMID: 32929218 DOI: 10.1038/s41418-020-00621-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 02/07/2023] Open
Abstract
The non-canonical inflammasome is an emerging crucial player in the development of inflammatory and neurodegenerative diseases. It is activated by direct sensing of cytosolic lipopolysaccharide (LPS) by caspase-11 (CASP11), which then induces pyroptosis, an inflammatory form of regulated cell death. Here, we report that tyrosine kinase 2 (TYK2), a cytokine receptor-associated kinase, is a critical upstream regulator of CASP11. Absence of TYK2 or its kinase activity impairs the transcriptional induction of CASP11 in vitro and in vivo and protects mice from LPS-induced lethality. Lack of TYK2 or its enzymatic activity inhibits macrophage pyroptosis and impairs release of mature IL-1β and IL-18 specifically in response to intracellular LPS. Deletion of TYK2 in myeloid cells reduces LPS-induced IL-1β and IL-18 production in vivo, highlighting the importance of these cells in the inflammatory response to LPS. In support of our data generated with genetically engineered mice, pharmacological inhibition of TYK2 reduced LPS-induced upregulation of CASP11 in bone marrow-derived macrophages (BMDMs) and of its homolog CASP5 in human macrophages. Our study provides insights into the regulation of CASP11 in vivo and uncovered a novel link between TYK2 activity and CASP11-dependent inflammation.
Collapse
|