Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.000
Filter
1.
Nat Rev Rheumatol ; 20(5): 258-271, 2024 May.
Article in English | MEDLINE | ID: mdl-38600215

ABSTRACT

In rheumatoid arthritis, juvenile idiopathic arthritis and other forms of inflammatory arthritis, the immune system targets certain joints but not others. The pattern of joints affected varies by disease and by individual, with flares most commonly involving joints that were previously inflamed. This phenomenon, termed joint-specific memory, is difficult to explain by systemic immunity alone. Mechanisms of joint-specific memory include the involvement of synovial resident memory T cells that remain in the joint during remission and initiate localized disease recurrence. In addition, arthritis-induced durable changes in synovial fibroblasts and macrophages can amplify inflammation in a site-specific manner. Together with ongoing systemic processes that promote extension of arthritis to new joints, these local factors set the stage for a stepwise progression in disease severity, a paradigm for arthritis chronicity that we term the joint accumulation model. Although durable drug-free remission through early treatment remains elusive for most forms of arthritis, the joint accumulation paradigm defines new therapeutic targets, emphasizes the importance of sustained treatment to prevent disease extension to new joints, and identifies a rolling window of opportunity for altering the natural history of arthritis that extends well beyond the initiation phase of disease.


Subject(s)
Arthritis, Rheumatoid , Memory T Cells , Humans , Memory T Cells/immunology , Arthritis, Rheumatoid/immunology , Joints/immunology , Joints/pathology , Immunologic Memory/immunology , Disease Progression , Animals , Synovial Membrane/immunology , Synovial Membrane/pathology , Arthritis/immunology
2.
J Virol ; 98(1): e0110223, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38169294

ABSTRACT

Mayaro virus (MAYV) is an emerging arbovirus member of the Togaviridae family and Alphavirus genus. MAYV infection causes an acute febrile illness accompanied by persistent polyarthralgia and myalgia. Understanding the mechanisms involved in arthritis caused by alphaviruses is necessary to develop specific therapies. In this work, we investigated the role of the CCL2/CCR2 axis in the pathogenesis of MAYV-induced disease. For this, wild-type (WT) C57BL/6J and CCR2-/- mice were infected with MAYV subcutaneously and evaluated for disease development. MAYV infection induced an acute inflammatory disease in WT mice. The immune response profile was characterized by an increase in the production of inflammatory mediators, such as IL-6, TNF, and CCL2. Higher levels of CCL2 at the local and systemic levels were followed by the significant recruitment of CCR2+ macrophages and a cellular response orchestrated by these cells. CCR2-/- mice showed an increase in CXCL-1 levels, followed by a replacement of the macrophage inflammatory infiltrate by neutrophils. Additionally, the absence of the CCR2 receptor protected mice from bone loss induced by MAYV. Accordingly, the silencing of CCL2 chemokine expression in vivo and the pharmacological blockade of CCR2 promoted a partial improvement in disease. Cell culture data support the mechanism underlying the bone pathology of MAYV, in which MAYV infection promotes a pro-osteoclastogenic microenvironment mediated by CCL2, IL-6, and TNF, which induces the migration and differentiation of osteoclast precursor cells. Overall, these data contribute to the understanding of the pathophysiology of MAYV infection and the identification future of specific therapeutic targets in MAYV-induced disease.IMPORTANCEThis work demonstrates the role of the CCL2/CCR2 axis in MAYV-induced disease. The infection of wild-type (WT) C57BL/6J and CCR2-/- mice was associated with high levels of CCL2, an important chemoattractant involved in the recruitment of macrophages, the main precursor of osteoclasts. In the absence of the CCR2 receptor, there is a mitigation of macrophage migration to the target organs of infection and protection of these mice against bone loss induced by MAYV infection. Much evidence has shown that host immune response factors contribute significantly to the tissue damage associated with alphavirus infections. Thus, this work highlights molecular and cellular targets involved in the pathogenesis of arthritis triggered by MAYV and identifies novel therapeutic possibilities directed to the host inflammatory response unleashed by MAYV.


Subject(s)
Alphavirus Infections , Arthritis , Chemokine CCL2 , Receptors, CCR2 , Animals , Mice , Alphavirus , Alphavirus Infections/immunology , Arthritis/immunology , Arthritis/virology , Chemokine CCL2/immunology , Interleukin-6/immunology , Mice, Inbred C57BL , Receptors, CCR2/immunology , Mice, Knockout , Male , Bone Diseases/virology
3.
Cell Rep ; 42(8): 113006, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37610870

ABSTRACT

Immune-suppressive effects of myeloid-derived suppressor cells (MDSCs) are well characterized during anti-tumor immunity. The complex mechanisms promoting MDSC development and their regulatory effects during autoimmune diseases are less understood. We demonstrate that the endogenous alarmin S100A8/A9 reprograms myeloid cells to a T cell suppressing phenotype during autoimmune arthritis. Treatment of myeloid precursors with S100-alarmins during differentiation induces MDSCs in a Toll-like receptor 4-dependent manner. Consequently, knockout of S100A8/A9 aggravates disease activity in collagen-induced arthritis due to a deficit of MDSCs in local lymph nodes, which could be corrected by adoptive transfer of S100-induced MDSCs. Blockade of MDSC function in vivo aggravates disease severity in arthritis. Therapeutic application of S100A8 induces MDSCs in vivo and suppresses the inflammatory phenotype of S100A9ko mice. Accordingly, the interplay of T cell-mediated autoimmunity with a defective innate immune regulation is crucial for autoimmune arthritis, which should be considered for future innovative therapeutic options.


Subject(s)
Arthritis , Calgranulin A , Calgranulin B , Myeloid-Derived Suppressor Cells , Animals , Mice , Arthritis/immunology , Arthritis/metabolism , Arthritis/pathology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Myeloid-Derived Suppressor Cells/cytology , Myeloid-Derived Suppressor Cells/immunology , Disease Models, Animal , Cell Differentiation , Nitric Oxide/metabolism , Signal Transduction , Toll-Like Receptor 4/metabolism , Calgranulin A/metabolism , Calgranulin B/metabolism
4.
Semin Arthritis Rheum ; 58: 152131, 2023 02.
Article in English | MEDLINE | ID: mdl-36527929

ABSTRACT

OBJECTIVES: To compare the magnitude of cognitive impairment against age-expected levels across the immune mediated inflammatory diseases (IMIDs: systemic lupus erythematosus [SLE], rheumatoid arthritis [RA], axial spondyloarthritis [axSpA], psoriatic arthritis [PsA], psoriasis [PsO]). METHODS: A pre-defined search strategy was implemented in Medline, Embase and Psychinfo on 29/05/2021. Inclusion criteria were: (i) observational studies of an IMID, (ii) healthy control comparison, (iii) measuring cognitive ability (overall, memory, complex attention/executive function, language/verbal fluency), and (iv) sufficient data for meta-analysis. Standardised mean differences (SMD) in cognitive assessments between IMIDs and controls were pooled using random-effects meta-analysis. IMIDs were compared using meta-regression. RESULTS: In total, 65 IMID groups were included (SLE: 39, RA: 19, axSpA: 1, PsA: 2 PsO: 4), comprising 3141 people with IMIDs and 9333 controls. People with IMIDs had impairments in overall cognition (SMD: -0.57 [95% CI -0.70, -0.43]), complex attention/executive function (SMD -0.57 [95% CI -0.69, -0.44]), memory (SMD -0.55 [95% CI -0.68, -0.43]) and language/verbal fluency (SMD -0.51 [95% CI -0.68, -0.34]). People with RA and people with SLE had similar magnitudes of cognitive impairment in relation to age-expected levels. People with neuropsychiatric SLE had larger impairment in overall cognition compared with RA. CONCLUSIONS: People with IMIDs have moderate impairments across a range of cognitive domains. People with RA and SLE have similar magnitudes of impairment against their respective age-expected levels, calling for greater recognition of cognitive impairment in both conditions. To further understand cognition in the IMIDs, more large-scale, longitudinal studies are needed.


Subject(s)
Arthritis , Cognitive Dysfunction , Lupus Erythematosus, Systemic , Psoriasis , Humans , Arthritis, Psoriatic/complications , Arthritis, Psoriatic/immunology , Arthritis, Rheumatoid/complications , Arthritis, Rheumatoid/immunology , Cognition , Cognitive Dysfunction/etiology , Cognitive Dysfunction/immunology , Lupus Erythematosus, Systemic/complications , Axial Spondyloarthritis/complications , Axial Spondyloarthritis/immunology , Arthritis/complications , Arthritis/immunology , Inflammation/complications , Inflammation/immunology
5.
J Virol ; 96(17): e0099922, 2022 09 14.
Article in English | MEDLINE | ID: mdl-36000846

ABSTRACT

Arthritogenic alphaviruses are mosquito-borne arboviruses that include several re-emerging human pathogens, including the chikungunya (CHIKV), Ross River (RRV), Mayaro (MAYV), and o'nyong-nyong (ONNV) virus. Arboviruses are transmitted via a mosquito bite to the skin. Herein, we describe intradermal RRV infection in a mouse model that replicates the arthritis and myositis seen in humans with Ross River virus disease (RRVD). We show that skin infection with RRV results in the recruitment of inflammatory monocytes and neutrophils, which together with dendritic cells migrate to draining lymph nodes (LN) of the skin. Neutrophils and monocytes are productively infected and traffic virus from the skin to LN. We show that viral envelope N-linked glycosylation is a key determinant of skin immune responses and disease severity. RRV grown in mammalian cells elicited robust early antiviral responses in the skin, while RRV grown in mosquito cells stimulated poorer early antiviral responses. We used glycan mass spectrometry to characterize the glycan profile of mosquito and mammalian cell-derived RRV, showing deglycosylation of the RRV E2 glycoprotein is associated with curtailed skin immune responses and reduced disease following intradermal infection. Altogether, our findings demonstrate skin infection with an arthritogenic alphavirus leads to musculoskeletal disease and envelope glycoprotein glycosylation shapes disease outcome. IMPORTANCE Arthritogenic alphaviruses are transmitted via mosquito bites through the skin, potentially causing debilitating diseases. Our understanding of how viral infection starts in the skin and how virus systemically disseminates to cause disease remains limited. Intradermal arbovirus infection described herein results in musculoskeletal pathology, which is dependent on viral envelope N-linked glycosylation. As such, intradermal infection route provides new insights into how arboviruses cause disease and could be extended to future investigations of skin immune responses following infection with other re-emerging arboviruses.


Subject(s)
Alphavirus Infections , Arthritis , Myositis , Polysaccharides , Ross River virus , Skin , Alphavirus Infections/complications , Alphavirus Infections/immunology , Animals , Antiviral Agents/immunology , Arthritis/complications , Arthritis/immunology , Culicidae/virology , Dendritic Cells , Disease Models, Animal , Glycosylation , Humans , Mass Spectrometry , Mice , Monocytes , Myositis/complications , Myositis/immunology , Neutrophils , Polysaccharides/chemistry , Polysaccharides/immunology , Ross River virus/immunology , Skin/immunology , Skin/virology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/immunology
6.
Nature ; 609(7926): 348-353, 2022 09.
Article in English | MEDLINE | ID: mdl-35978195

ABSTRACT

The mammalian immune system uses various pattern recognition receptors to recognize invaders and host damage and transmits this information to downstream immunometabolic signalling outcomes. Laccase domain-containing 1 (LACC1) protein is an enzyme highly expressed in inflammatory macrophages and serves a central regulatory role in multiple inflammatory diseases such as inflammatory bowel diseases, arthritis and clearance of microbial infection1-4. However, the biochemical roles required for LACC1 functions remain largely undefined. Here we elucidated a shared biochemical function of LACC1 in mice and humans, converting L-citrulline to L-ornithine (L-Orn) and isocyanic acid and serving as a bridge between proinflammatory nitric oxide synthase (NOS2) and polyamine immunometabolism. We validated the genetic and mechanistic connections among NOS2, LACC1 and ornithine decarboxylase 1 (ODC1) in mouse models and bone marrow-derived macrophages infected by Salmonella enterica Typhimurium. Strikingly, LACC1 phenotypes required upstream NOS2 and downstream ODC1, and Lacc1-/- chemical complementation with its product L-Orn significantly restored wild-type activities. Our findings illuminate a previously unidentified pathway in inflammatory macrophages, explain why its deficiency may contribute to human inflammatory diseases and suggest that L-Orn could serve as a nutraceutical to ameliorate LACC1-associated immunological dysfunctions such as arthritis or inflammatory bowel disease.


Subject(s)
Inflammation , Intracellular Signaling Peptides and Proteins , Macrophages , Nitric Oxide Synthase Type II , Animals , Arthritis/immunology , Arthritis/metabolism , Citrulline/metabolism , Cyanates/metabolism , Humans , Inflammation/enzymology , Inflammation/immunology , Inflammation/metabolism , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Nitric Oxide Synthase Type II/metabolism , Ornithine/metabolism , Ornithine Decarboxylase/metabolism , Polyamines/metabolism , Salmonella typhimurium/immunology
7.
Int J Mol Sci ; 23(3)2022 Jan 20.
Article in English | MEDLINE | ID: mdl-35163028

ABSTRACT

While type 2 immunity has traditionally been associated with the control of parasitic infections and allergic reactions, increasing evidence suggests that type 2 immunity exerts regulatory functions on inflammatory diseases such as arthritis, and also on bone homeostasis. This review summarizes the current evidence of the regulatory role of type 2 immunity in arthritis and bone. Key type 2 cytokines, like interleukin (IL)-4 and IL-13, but also others such as IL-5, IL-9, IL-25, and IL-33, exert regulatory properties on arthritis, dampening inflammation and inducing resolution of joint swelling. Furthermore, these cytokines share anti-osteoclastogenic properties and thereby reduce bone resorption and protect bone. Cellular effectors of this action are both T cells (i.e., Th2 and Th9 cells), but also non-T cells, like type 2 innate lymphoid cells (ILC2). Key regulatory actions mediated by type 2 cytokines and immune cells on both inflammation as well as bone homeostasis are discussed.


Subject(s)
Adaptive Immunity , Arthritis/immunology , Bone and Bones/immunology , Immunity, Innate/immunology , Inflammation/immunology , Th2 Cells/immunology , Animals , Arthritis/pathology , Bone and Bones/pathology , Humans , Inflammation/pathology
8.
JCI Insight ; 7(2)2022 01 25.
Article in English | MEDLINE | ID: mdl-35076027

ABSTRACT

Secreted phospholipase A2-IIA (sPLA2-IIA) hydrolyzes phospholipids to liberate lysophospholipids and fatty acids. Given its poor activity toward eukaryotic cell membranes, its role in the generation of proinflammatory lipid mediators is unclear. Conversely, sPLA2-IIA efficiently hydrolyzes bacterial membranes. Here, we show that sPLA2-IIA affects the immune system by acting on the intestinal microbial flora. Using mice overexpressing transgene-driven human sPLA2-IIA, we found that the intestinal microbiota was critical for both induction of an immune phenotype and promotion of inflammatory arthritis. The expression of sPLA2-IIA led to alterations of the intestinal microbiota composition, but housing in a more stringent pathogen-free facility revealed that its expression could affect the immune system in the absence of changes to the composition of this flora. In contrast, untargeted lipidomic analysis focusing on bacteria-derived lipid mediators revealed that sPLA2-IIA could profoundly alter the fecal lipidome. The data suggest that a singular protein, sPLA2-IIA, produces systemic effects on the immune system through its activity on the microbiota and its lipidome.


Subject(s)
Arthritis , Bacterial Physiological Phenomena/immunology , Gastrointestinal Microbiome/physiology , Group II Phospholipases A2/metabolism , Lipid Metabolism/immunology , Animals , Animals, Genetically Modified , Arthritis/immunology , Arthritis/microbiology , Humans , Immune System Phenomena , Lipidomics/methods , Mice , Models, Animal , Pathology, Molecular/methods , Transgenes
9.
Protein Expr Purif ; 189: 105966, 2022 01.
Article in English | MEDLINE | ID: mdl-34627999

ABSTRACT

Nerve growth factor (NGF) is produced and released in injured tissues or chronic pain tissues caused by other diseases. Studies have shown that monoclonal antibodies targeting NGF have a good efficacy in the treatment of osteoarthritis (OA), low back pain and chronic pain, which may be a promising therapy. In this study, DNA sequences of NGF-his and NGF-hFc were synthesized using eukaryotic expression system and subcloned into pTT5 expression vector. After that, NGF proteins were expressed by transient expression in HEK293E cells. We immunized mice with NGF-hFc protein and fused mouse spleen cells to prepare hybridomas. NGF-His protein was used to screen out the hybridoma supernatant that could directly bind to NGF. Antibodies were purified from hybridioma supernatant. Futhermore, via surface plasmon resonance (SPR) screening, six anti-NGF mAbs were screened to block the binding of NGF and TrkA receptor in the treatment of chronic pain. Among them, 58F10G10H showed high affinity (KD = 1.03 × 10-9 M) and even better than that of positive control antibody Tanezumab (KD = 1.53 × 10-9 M). Moreover, the specific reactivity of 58F10G10H was demonstrated by TF-1 cell proliferation activity experiments, competitive binding Enzyme-linked immunosorbent assay (ELISA) and the arthritis animal models in mice, respectively. In conclusion, in this study, a method for the preparation of high-yield NGF-HFC and NGF-His proteins was designed, and a high-affinity monoclonal antibody against NGF with potential for basic research and clinical application was prepared.


Subject(s)
Antibodies, Monoclonal/pharmacology , Arthritis/drug therapy , Nerve Growth Factor/antagonists & inhibitors , Pain/prevention & control , Receptor, trkA/antagonists & inhibitors , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal, Humanized/pharmacology , Antibody Affinity , Antibody Specificity , Arthritis/genetics , Arthritis/immunology , Arthritis/pathology , Disease Models, Animal , Female , Gene Expression , HEK293 Cells , Humans , Hybridomas/chemistry , Hybridomas/immunology , Immunization , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/immunology , Lymphocytes/chemistry , Lymphocytes/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nerve Growth Factor/genetics , Nerve Growth Factor/immunology , Pain/genetics , Pain/immunology , Pain/pathology , Receptor, trkA/genetics , Receptor, trkA/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology
10.
J Allergy Clin Immunol ; 149(1): 176-188.e7, 2022 01.
Article in English | MEDLINE | ID: mdl-34175136

ABSTRACT

BACKGROUND: Blau syndrome (BS) is an autoinflammatory disease associated with mutations in nucleotide-binding oligomerization domain 2. Although treatments with anti-TNF agents have been reported to be effective, the underlying molecular mechanisms remain unclear. OBJECTIVE: We aimed to elucidate the mechanisms of autoinflammation in patients with BS and to clarify how anti-TNF treatment controls the disease phenotype at the cellular level in clinical samples. METHODS: Macrophages were differentiated from monocytes of 7 BS patients, and global transcriptional profiles of 5 patients were analyzed with or without IFN-γ stimulation. Macrophages were also generated from BS-specific induced pluripotent stem cells (iPSCs), and their transcriptome was examined for comparison. RESULTS: Aberrant inflammatory responses were observed upon IFN-γ stimulation in macrophages from untreated BS patients, but not in those from patients treated with anti-TNF. iPSC-derived macrophages carrying a disease-associated mutation also showed IFN-γ-dependent accelerated inflammatory responses. Comparisons of peripheral blood- and iPSC-derived macrophages revealed the upregulation of nuclear factor kappa-light-chain enhancer of activated B cells (NF-κB) targets in unstimulated macrophages as a common feature. CONCLUSIONS: IFN-γ stimulation is one of the key signals driving aberrant inflammatory responses in BS-associated macrophages. However, long-term treatment with anti-TNF agents ameliorates such abnormalities even in the presence of IFN-γ stimulation. Our data thus suggest that preexposure to TNF or functionally similar cytokines inducing NF-κB-driven proinflammatory signaling during macrophage development is a prerequisite for accelerated inflammatory responses upon IFN-γ stimulation in BS.


Subject(s)
Arthritis/immunology , Interferon-gamma/immunology , Macrophages/immunology , Sarcoidosis/immunology , Synovitis/immunology , Tumor Necrosis Factor Inhibitors/pharmacology , Uveitis/immunology , Adult , Arthritis/drug therapy , Arthritis/genetics , Cell Line , Child , Child, Preschool , Female , Humans , Induced Pluripotent Stem Cells/cytology , Male , NF-kappa B/immunology , Sarcoidosis/drug therapy , Sarcoidosis/genetics , Synovitis/drug therapy , Synovitis/genetics , Transcriptome , Tumor Necrosis Factor Inhibitors/therapeutic use , Uveitis/drug therapy , Uveitis/genetics , Young Adult
11.
J Immunol ; 208(3): 571-581, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34965962

ABSTRACT

IDO2 is one of two closely related tryptophan catabolizing enzymes induced under inflammatory conditions. In contrast to the immunoregulatory role defined for IDO1 in cancer models, IDO2 has a proinflammatory function in models of autoimmunity and contact hypersensitivity. In humans, two common single-nucleotide polymorphisms have been identified that severely impair IDO2 enzymatic function, such that <25% of individuals express IDO2 with full catalytic potential. This, together with IDO2's relatively weak enzymatic activity, suggests that IDO2 may have a role outside of its function in tryptophan catabolism. To determine whether the enzymatic activity of IDO2 is required for its proinflammatory function, we used newly generated catalytically inactive IDO2 knock-in mice together with established models of contact hypersensitivity and autoimmune arthritis. Contact hypersensitivity was attenuated in catalytically inactive IDO2 knock-in mice. In contrast, induction of autoimmune arthritis was unaffected by the absence of IDO2 enzymatic activity. In pursuing this nonenzymatic IDO2 function, we identified GAPDH, Runx1, RANbp10, and Mgea5 as IDO2-binding proteins that do not interact with IDO1, implicating them as potential mediators of IDO2-specific function. Taken together, our findings identify a novel function for IDO2, independent of its tryptophan catabolizing activity, and suggest that this nonenzymatic function could involve multiple signaling pathways. These data show that the enzymatic activity of IDO2 is required only for some inflammatory immune responses and provide, to our knowledge, the first evidence of a nonenzymatic role for IDO2 in mediating autoimmune disease.


Subject(s)
Arthritis/immunology , Autoimmunity/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Animals , Antigens, Neoplasm/metabolism , Cell Line , Core Binding Factor Alpha 2 Subunit/metabolism , Gene Knock-In Techniques , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , Humans , Inflammation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Polymorphism, Single Nucleotide/genetics
12.
Int J Mol Sci ; 22(23)2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34884486

ABSTRACT

Lysophosphatidylserine (lysoPS) is known to regulate immune cell functions. Phospholipase A1 member A (PLA1A) can generate this bioactive lipid through hydrolysis of sn-1 fatty acids on phosphatidylserine (PS). PLA1A has been associated with cancer metastasis, asthma, as well as acute coronary syndrome. However, the functions of PLA1A in the development of systemic autoimmune rheumatic diseases remain elusive. To investigate the possible implication of PLA1A during rheumatic diseases, we monitored PLA1A in synovial fluids from patients with rheumatoid arthritis and plasma of early-diagnosed arthritis (EA) patients and clinically stable systemic lupus erythematosus (SLE) patients. We used human primary fibroblast-like synoviocytes (FLSs) to evaluate the PLA1A-induced biological responses. Our results highlighted that the plasma concentrations of PLA1A in EA and SLE patients were elevated compared to healthy donors. High concentrations of PLA1A were also detected in synovial fluids from rheumatoid arthritis patients compared to those from osteoarthritis (OA) and gout patients. The origin of PLA1A in FLSs and the arthritic joints remained unknown, as healthy human primary FLSs does not express the PLA1A transcript. Besides, the addition of recombinant PLA1A stimulated cultured human primary FLSs to secrete IL-8. Preincubation with heparin, autotaxin (ATX) inhibitor HA130 or lysophosphatidic acid (LPA) receptor antagonist Ki16425 reduced PLA1A-induced-secretion of IL-8. Our data suggested that FLS-associated PLA1A cleaves membrane-exposed PS into lysoPS, which is subsequently converted to LPA by ATX. Since primary FLSs do not express any lysoPS receptors, the data suggested PLA1A-mediated pro-inflammatory responses through the ATX-LPA receptor signaling axis.


Subject(s)
Arthritis/pathology , Fibroblasts/pathology , Gout/pathology , Lupus Erythematosus, Systemic/pathology , Phospholipases A1/metabolism , Phosphoric Diester Hydrolases/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Synoviocytes/pathology , Arthritis/genetics , Arthritis/immunology , Arthritis/metabolism , Case-Control Studies , Female , Fibroblasts/immunology , Fibroblasts/metabolism , Gout/genetics , Gout/immunology , Gout/metabolism , Humans , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Male , Phospholipases A1/genetics , Phosphoric Diester Hydrolases/genetics , Receptors, Lysophosphatidic Acid/genetics , Synovial Fluid/immunology , Synovial Fluid/metabolism , Synoviocytes/immunology , Synoviocytes/metabolism
13.
Nat Rev Rheumatol ; 17(11): 651-664, 2021 11.
Article in English | MEDLINE | ID: mdl-34599320

ABSTRACT

Fatigue is a complex phenomenon and an important health concern for many people with chronic inflammatory rheumatic diseases, such as rheumatoid arthritis, psoriatic arthritis, primary Sjögren syndrome and systemic lupus erythematosus. Although some clinical trials have shown the benefits of cognitive behavioural therapy in fatigue management, the effect of this approach is relatively modest, and no curative treatment has been identified. The pathogenesis of fatigue remains unclear. Despite many challenges and limitations, a growing body of research points to roles for the immune system, the central and autonomic nervous systems and the neuroendocrine system in the induction and maintenance of fatigue in chronic diseases. New insights indicate that sleep, genetic susceptibility, metabolic disturbances and other biological and physiological mechanisms contribute to fatigue. Furthermore, understanding of the relationships between psychosocial factors and fatigue is increasing. However, the interrelationships between these diverse mechanisms and fatigue remain poorly defined. In this Review, we outline various biological, physiological and psychosocial determinants of fatigue in inflammatory rheumatic diseases, and propose mechanistic and conceptual models of fatigue to summarize current understanding, stimulate debate and develop further research ideas.


Subject(s)
Arthritis , Connective Tissue Diseases , Fatigue , Rheumatic Diseases , Vasculitis , Arthritis/immunology , Biomedical Research , Chronic Disease , Connective Tissue Diseases/immunology , Fatigue/diagnosis , Fatigue/etiology , Fatigue/physiopathology , Fatigue/therapy , Forecasting , Humans , Inflammation/immunology , Rheumatic Diseases/complications , Rheumatic Diseases/immunology , Vasculitis/immunology
14.
Rheumatology (Oxford) ; 60(Suppl 4): iv4-iv15, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34668017

ABSTRACT

IL-23 is a cytokine member of the IL-12 superfamily. These heterodimeric cytokines offer broad immune regulatory activity with potential effector function in inflammatory arthritis. IL-23 is a pro-inflammatory cytokine secreted by dendritic cells and macrophages. It plays a key role in both innate and adaptive immunity. By promoting and maintaining T cell differentiation into Th17 T cells, IL-23 is a key player in the pathogenesis of rheumatic diseases. Data from pre-clinical IL-23 knockout models show the major importance of IL-23 in development of arthritis. The induction and maintenance of type 17 cells, which secrete IL-17A and other pro-inflammatory cytokines, contributes to local synovial inflammation and skin inflammation in PsA, and perhaps in RA. Commensurate with this, therapeutic strategies targeting IL-23 have proven efficient in PsA in several studies, albeit not yet in RA.


Subject(s)
Arthritis/immunology , Interleukin-23/metabolism , Animals , Arthritis/drug therapy , Arthritis/metabolism , Humans , Interleukin-23/antagonists & inhibitors , Molecular Targeted Therapy
15.
Ann Rheum Dis ; 80(12): 1604-1614, 2021 12.
Article in English | MEDLINE | ID: mdl-34663597

ABSTRACT

Crystal structures activate innate immune cells, especially macrophages and initiate inflammatory responses. We aimed to understand the role of the mechanosensitive TRPV4 channel in crystal-induced inflammation. Real-time RT-PCR, RNAscope in situ hybridisation, and Trpv4eGFP mice were used to examine TRPV4 expression and whole-cell patch-clamp recording and live-cell Ca2+ imaging were used to study TRPV4 function in mouse synovial macrophages and human peripheral blood mononuclear cells (PBMCs). Both genetic deletion and pharmacological inhibition approaches were used to investigate the role of TRPV4 in NLRP3 inflammasome activation induced by diverse crystals in vitro and in mouse models of crystal-induced pain and inflammation in vivo. TRPV4 was functionally expressed by synovial macrophages and human PBMCs and TRPV4 expression was upregulated by stimulation with monosodium urate (MSU) crystals and in human PBMCs from patients with acute gout flares. MSU crystal-induced gouty arthritis were significantly reduced by either genetic ablation or pharmacological inhibition of TRPV4 function. Mechanistically, TRPV4 mediated the activation of NLRP3 inflammasome by diverse crystalline materials but not non-crystalline NLRP3 inflammasome activators, driving the production of inflammatory cytokine interleukin-1ß which elicited TRPV4-dependent inflammatory responses in vivo. Moreover, chemical ablation of the TRPV1-expressing nociceptors significantly attenuated the MSU crystal-induced gouty arthritis. In conclusion, TRPV4 is a common mediator of inflammatory responses induced by diverse crystals through NLRP3 inflammasome activation in macrophages. TRPV4-expressing resident macrophages are critically involved in MSU crystal-induced gouty arthritis. A neuroimmune interaction between the TRPV1-expressing nociceptors and the TRPV4-expressing synovial macrophages contributes to the generation of acute gout flares.


Subject(s)
Arthralgia/metabolism , Arthritis/metabolism , Crystal Arthropathies/metabolism , Leukocytes, Mononuclear/metabolism , Macrophages/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Nociceptors/metabolism , TRPV Cation Channels/genetics , Adult , Animals , Arthralgia/immunology , Arthritis/immunology , Arthritis, Gouty/immunology , Arthritis, Gouty/metabolism , Crystal Arthropathies/immunology , Gout/immunology , Gout/metabolism , Humans , Inflammasomes/immunology , Inflammation , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Leukocytes, Mononuclear/immunology , Macrophages/immunology , Male , Mice , Middle Aged , Optical Imaging , Patch-Clamp Techniques , Synovial Membrane/cytology , THP-1 Cells , TRPV Cation Channels/agonists , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/metabolism , Uric Acid
17.
Clin Epigenetics ; 13(1): 192, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34663428

ABSTRACT

To define the role of SETD2 in the WNT5a signaling in the context of osteoclastogenesis, we exploited two different models: in vitro osteoclast differentiation, and K/BxN serum-induced arthritis model. We found that SETD2 and WNT5a were upregulated during osteoclast differentiation and after induction of arthritis. Using gain- and loss-of-function approaches in the myeloid cell, we confirmed that SETD2 regulated the osteoclast markers, and WNT5a via modulating active histone marks by enriching H3K36me3, and by reducing repressive H3K27me3 mark. Additionally, during osteoclastic differentiation, the transcription of Wnt5a was also associated with the active histone H3K9 and H4K8 acetylations. Mechanistically, SETD2 directed induction of NF-κß expression facilitated the recruitment of H3K9Ac and H4K8Ac around the TSS region of the Wnt5a gene, thereby, assisting osteoclast differentiation. Together these findings for the first time revealed that SETD2 mediated epigenetic regulation of Wnt5a plays a critical role in osteoclastogenesis and induced arthritis. Model for the Role of SETD2 dependent regulation of osteoclastic differentiation. A In monocyte cells SETD2-dependent H3K36 trimethylation help to create open chromatin region along with active enhancer mark, H3K27Ac. This chromatin state facilitated the loss of a suppressive H3K27me3 mark. B Additionally, SETD2 mediated induction of NF-κß expression leads to the recruitment of histone acetyl transferases, P300/PCAF, to the Wnt5a gene and establish H3K9Ac and H4K8Ac marks. Along with other activation marks, these acetylation marks help in Wnt5a transcription which leads to osteoclastogenesis.


Subject(s)
Epigenesis, Genetic/genetics , Histone-Lysine N-Methyltransferase/pharmacology , Osteogenesis/genetics , Wnt-5a Protein/adverse effects , Animals , Arthritis/immunology , Arthritis/physiopathology , Disease Models, Animal , Histone-Lysine N-Methyltransferase/genetics , Mice , Mice, Inbred C57BL , Osteogenesis/physiology , Transcriptional Activation/genetics , Wnt-5a Protein/genetics
18.
Sci Rep ; 11(1): 18679, 2021 09 21.
Article in English | MEDLINE | ID: mdl-34548542

ABSTRACT

Programmed necrosis, such as necroptosis and pyroptosis, is a highly pro-inflammatory cellular event that is associated with chronic inflammation. Although there are various triggers of pyroptosis and necroptosis in autoimmune tissue inflammation and subsequent lytic forms of cell death release abundant inflammatory mediators, including damage-associated molecular patterns and IL-1ß, capable of amplifying autoimmune Th17 effector functions, it remains largely unclear whether the programs play a crucial role in the pathogenesis of autoimmune arthritis. We herein report that Gasdermin D (Gsdmd) and receptor interacting serine/threonine kinase 3 (Ripk3)-key molecules of pyroptosis and necroptosis, respectively-are upregulated in inflamed synovial tissues, but dispensable for IL-1ß production and the development of IL-17-producing T helper (Th17) cell-mediated autoimmune arthritis in SKG mice. Gsdmd-/-, Ripk3-/-, or Gsdmd-/- Ripk3-/- SKG mice showed severe arthritis with expansion of arthritogenic Th17 cells in the draining LNs and inflamed joints, which was comparable to that in wild-type SKG mice. Despite the marked reduction of IL-1ß secretion from Gsdmd-/- or Ripk3-/- bone marrow-derived DCs by canonical stimuli, IL-1ß levels in the inflamed synovium were not affected in the absence of Gsdmd or Ripk3. Our results revealed that T cell-mediated autoimmune arthritis proceeds independently of the pyroptosis and necroptosis pathways.


Subject(s)
Arthritis/immunology , Autoimmune Diseases/immunology , Inflammation/immunology , Intracellular Signaling Peptides and Proteins/physiology , Phosphate-Binding Proteins/physiology , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Th17 Cells/immunology , Animals , Chronic Disease , Mice
19.
Cells ; 10(9)2021 09 21.
Article in English | MEDLINE | ID: mdl-34572149

ABSTRACT

The inflammatory cytokine interleukin-26 (IL-26) is highly expressed in the serum and synovial fluid of patients with inflammatory arthritis. The effect of IL-26 on human articular chondrocytes (HACs) remains unclear. Obesity is associated with disability of patients with rheumatoid arthritis and disease activity in those with ankylosing spondylitis. The saturated free fatty acid palmitate with IL-1ß can synergistically induce catabolic effects in HACs. The aim of this study was to evaluate the effects of IL-26 and palmitate in HACs. In this study, palmitate markedly synergizes the IL-26-induced proinflammatory effects and matrix protease, including COX-2, IL-6, and MMP-1, in HACs via the toll-like receptor 4 (TLR4)-ERK1/2-c-Jun signal transduction pathway. The synergistic catabolic effects of palmitate and IL-26 were attenuated by inhibitors of TLR4 (TAK242), ERK1/2 (U0126), or c-Jun (SP600125) in HACs and cartilage matrix. In addition, metformin, a potential inhibitor of TLR4, also decreased expression of COX-2 and IL-6 induced by co-incubation with IL-26 and palmitate. IL-26 and palmitate synergistically induced expression of inflammatory and catabolic mediators, resulting in articular cartilage matrix breakdown. The present study also revealed a possible mechanism and therapeutic targets against articular cartilage degradation by increased saturated fatty acids in patients with inflammatory arthritis.


Subject(s)
Chondrocytes/metabolism , Interleukins/metabolism , Palmitates/metabolism , Arthritis/immunology , Arthritis/metabolism , Arthritis/physiopathology , Arthritis, Rheumatoid/metabolism , Cartilage, Articular/metabolism , Chondrocytes/physiology , Genes, jun/physiology , Humans , Interleukins/immunology , MAP Kinase Signaling System/physiology , Metabolism/physiology , Osteoarthritis/metabolism , Signal Transduction/genetics , Synovial Membrane/metabolism , Taiwan , Toll-Like Receptor 4/metabolism
20.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Article in English | MEDLINE | ID: mdl-34507983

ABSTRACT

Arthritogenic alphaviruses are globally distributed, mosquito-transmitted viruses that cause rheumatological disease in humans and include Chikungunya virus (CHIKV), Mayaro virus (MAYV), and others. Although serological evidence suggests that some antibody-mediated heterologous immunity may be afforded by alphavirus infection, the extent to which broadly neutralizing antibodies that protect against multiple arthritogenic alphaviruses are elicited during natural infection remains unknown. Here, we describe the isolation and characterization of MAYV-reactive alphavirus monoclonal antibodies (mAbs) from a CHIKV-convalescent donor. We characterized 33 human mAbs that cross-reacted with CHIKV and MAYV and engaged multiple epitopes on the E1 and E2 glycoproteins. We identified five mAbs that target distinct regions of the B domain of E2 and potently neutralize multiple alphaviruses with differential breadth of inhibition. These broadly neutralizing mAbs (bNAbs) contain few somatic mutations and inferred germline-revertants retained neutralizing capacity. Two bNAbs, DC2.M16 and DC2.M357, protected against both CHIKV- and MAYV-induced musculoskeletal disease in mice. These findings enhance our understanding of the cross-reactive and cross-protective antibody response to human alphavirus infections.


Subject(s)
Alphavirus Infections/immunology , Antibodies, Monoclonal/isolation & purification , Broadly Neutralizing Antibodies/immunology , Alphavirus/immunology , Alphavirus/pathogenicity , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Arthritis/etiology , Arthritis/immunology , Arthritis/virology , Broadly Neutralizing Antibodies/isolation & purification , Broadly Neutralizing Antibodies/pharmacology , Chikungunya Fever/virology , Chikungunya virus/immunology , Chikungunya virus/pathogenicity , Cross Reactions , Epitopes/immunology , Germ Cells/immunology , Glycoproteins/immunology , Humans , Male , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...