Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
EMBO Mol Med ; 15(10): e17691, 2023 Oct 11.
Article in English | MEDLINE | ID: mdl-37694693

ABSTRACT

Arthritis is the most common extra-intestinal complication in inflammatory bowel disease (IBD). Conversely, arthritis patients are at risk for developing IBD and often display subclinical gut inflammation. These observations suggest a shared disease etiology, commonly termed "the gut-joint-axis." The clinical association between gut and joint inflammation is further supported by the success of common therapeutic strategies and microbiota dysbiosis in both conditions. Most data, however, support a correlative relationship between gut and joint inflammation, while causative evidence is lacking. Using two independent transgenic mouse arthritis models, either TNF- or IL-1ß dependent, we demonstrate that arthritis develops independently of the microbiota and intestinal inflammation, since both lines develop full-blown articular inflammation under germ-free conditions. In contrast, TNF-driven gut inflammation is fully rescued in germ-free conditions, indicating that the microbiota is driving TNF-induced gut inflammation. Together, our study demonstrates that although common inflammatory pathways may drive both gut and joint inflammation, the molecular triggers initiating such pathways are distinct in these tissues.

2.
Rheumatology (Oxford) ; 62(9): 3169-3178, 2023 09 01.
Article in English | MEDLINE | ID: mdl-36661300

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.


Subject(s)
Arthritis, Experimental , Arthritis, Psoriatic , Mice , Animals , Interleukin-17/metabolism , Arthritis, Psoriatic/drug therapy , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , X-Ray Microtomography , Inflammation/pathology , Cytokines , Interleukin-23/metabolism
3.
Adv Healthc Mater ; 12(8): e2201726, 2023 03.
Article in English | MEDLINE | ID: mdl-36468909

ABSTRACT

This is the first comprehensive study of the impact of biodegradation on the structure, surface potential, mechanical and piezoelectric properties of poly(3-hydroxybutyrate) (PHB) scaffolds supplemented with reduced graphene oxide (rGO) as well as cell behavior under static and dynamic mechanical conditions. There is no effect of the rGO addition up to 1.0 wt% on the rate of enzymatic biodegradation of PHB scaffolds for 30 d. The biodegradation of scaffolds leads to the depolymerization of the amorphous phase, resulting in an increase in the degree of crystallinity. Because of more regular dipole order in the crystalline phase, surface potential of all fibers increases after the biodegradation, with a maximum (361 ± 5 mV) after the addition of 1 wt% rGO into PHB as compared to pristine PHB fibers. By contrast, PHB-0.7rGO fibers manifest the strongest effective vertical (0.59 ± 0.03 pm V-1 ) and lateral (1.06 ± 0.02 pm V-1 ) piezoresponse owing to a greater presence of electroactive ß-phase. In vitro assays involving primary human fibroblasts reveal equal biocompatibility and faster cell proliferation on PHB-0.7rGO scaffolds compared to pure PHB and nonpiezoelectric polycaprolactone scaffolds. Thus, the developed biodegradable PHB-rGO scaffolds with enhanced piezoresponse are promising for tissue-engineering applications.


Subject(s)
Hydroxybutyrates , Tissue Scaffolds , Humans , Tissue Scaffolds/chemistry , 3-Hydroxybutyric Acid , Hydroxybutyrates/chemistry , Tissue Engineering/methods , Polyesters/chemistry
4.
Sci Transl Med ; 13(616): eabg1210, 2021 Oct 20.
Article in English | MEDLINE | ID: mdl-34669443

ABSTRACT

Spondyloarthritis (SpA), a type 3 immunity-mediated inflammatory arthritis, is a systemic rheumatic disease that primarily affects the joints, spine, gut, skin, and eyes. Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine, yet MIF's pathological role in SpA is unknown. Here, we observed that the expression of MIF and its receptor CD74 is increased in blood and tissues of curdlan (ß-glucan)­treated SKG mice, a mouse model of SpA. We found that neutrophils substantially expanded and produced MIF in curdlan-treated SKG mice and that human neutrophils from SpA patients secreted higher concentrations of MIF compared to healthy individuals. Although genetic deletion of Mif (Mif−/−) substantially suppressed the severity of SpA features, adoptive transfer of inflammatory neutrophils induced SpA pathology in curdlan-treated Mif−/− SKG mice; in contrast, blocking the function of neutrophils with anti­Gr-1 antibody suppressed the curdlan-induced SpA-like phenotype. We also determined that systemic MIF overexpression was sufficient to induce SpA-like clinical features in SKG mice with enhanced type 3 immunity, whereas SKG mice treated with a MIF antagonist prevented or attenuated curdlan-induced SpA manifestations. Mechanistically, we identified that MIF intensifies type 3 immunity by boosting human and mouse T regulatory cell (Treg) acquisition of a TH17 cell­like phenotype, including the up-regulation of interleukin-17 (IL-17) and IL-22 in vitro. Tregs in blood and synovial fluids from SpA patients have a pathologic TH17 phenotype. These results indicate that MIF is a crucial regulator and a potential therapeutic target in type 3 immunity-mediated arthritis.


Subject(s)
Macrophage Migration-Inhibitory Factors , Spondylarthritis , Animals , Disease Models, Animal , Humans , Macrophage Migration-Inhibitory Factors/genetics , Mice
5.
Front Genet ; 12: 685280, 2021.
Article in English | MEDLINE | ID: mdl-34290741

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.

6.
Rheumatology (Oxford) ; 60(Suppl 4): iv16-iv27, 2021 10 19.
Article in English | MEDLINE | ID: mdl-33961030

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.


Subject(s)
Interleukin-23/physiology , Spondylarthritis/immunology , Animals , Bacterial Translocation , Humans , Joints/immunology , Psoriasis/etiology , Receptors, Interleukin/metabolism , Skin/metabolism , Spondylarthritis/microbiology
8.
J Immunol ; 205(12): 3300-3310, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33199537

ABSTRACT

Gout is a painful arthritic inflammatory disease caused by buildup of monosodium urate (MSU) crystals in the joints. Colchicine, a microtubule-depolymerizing agent that is used in prophylaxis and treatment of acute gout flare, alleviates the painful inflammatory response to MSU crystals. Using i.p. and intra-articular mouse models of gout-like inflammation, we found that GEF-H1/GEF-H1/AHRGEF2, a microtubule-associated Rho-GEF, was necessary for the inhibitory effect of colchicine on neutrophil recruitment. GEF-H1 was required for neutrophil polarization in response to colchicine, characterized by uropod formation, accumulation of F-actin and myosin L chain at the leading edge, and accumulation of phosphorylated myosin L chain, flotillin-2, and P-selectin glycoprotein ligand-1 (PSGL-1) in the uropod. Wild-type neutrophils that were pre-exposed to colchicine failed to roll or accumulate on activated endothelial monolayers, whereas GEF-H1 knockout (GEF-H1-/-) neutrophils were unaffected by treatment with colchicine. In vivo, colchicine blocked MSU-induced recruitment of neutrophils to the peritoneum and the synovium in wild-type mice, but not in GEF-H1-/- mice. Inhibition of macrophage IL-1ß production by colchicine was independent of GEF-H1, supporting a neutrophil-intrinsic mode of action. Our results suggest that the anti-inflammatory effects of colchicine in acute gout-like inflammation can be accounted for by inhibition of neutrophil-rolling interactions with the inflamed vasculature and occurs through GEF-H1-dependent neutrophil stimulation by colchicine. These results contribute to our understanding of the therapeutic action of colchicine, and could inform the application of this drug in other conditions.


Subject(s)
Colchicine/pharmacology , Gout , Leukocyte Rolling , Neutrophil Infiltration/drug effects , Neutrophils , Rho Guanine Nucleotide Exchange Factors/immunology , Actins/genetics , Actins/immunology , Animals , Disease Models, Animal , Gout/drug therapy , Gout/genetics , Gout/immunology , Gout/pathology , Leukocyte Rolling/drug effects , Leukocyte Rolling/genetics , Leukocyte Rolling/immunology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Knockout , Myosin Light Chains , Neutrophils/immunology , Neutrophils/pathology , Rho Guanine Nucleotide Exchange Factors/genetics
9.
Nat Rev Rheumatol ; 16(9): 536, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32733005

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

10.
Nat Rev Rheumatol ; 16(8): 415-433, 2020 08.
Article in English | MEDLINE | ID: mdl-32661321

ABSTRACT

Gut inflammation is strongly associated with spondyloarthritis (SpA), as exemplified by the high prevalence of inflammatory bowel disease (IBD) and the even higher occurrence of subclinical gut inflammation in patients with SpA. The gut-joint axis of inflammation in SpA is further reinforced by similarities in immunopathogenesis at both anatomical sites and by the clinical success of therapies blocking TNF and IL-23 in IBD and in some forms of SpA. Many genetic risk factors are shared between SpA and IBD, and changes in the composition of gut microbiota are seen in both diseases. Current dogma is that inflammation in SpA initiates in the gut and leads to joint inflammation; however, although conceptually attractive, some research does not support this causal relationship. For example, therapies targeting IL-17A are efficacious in the joint but not the gut, and interfering with gut trafficking by targeting molecules such as α4ß7 in IBD can lead to onset or flares of SpA. Several important knowledge gaps remain that must be addressed in future studies. Determining the true nature of the gut-joint axis has real-world implications for the treatment of patients with co-incident IBD and SpA and for the repurposing of therapeutics from one disease to the other.


Subject(s)
Gastrointestinal Microbiome , Inflammatory Bowel Diseases/complications , Spondylarthritis/etiology , Animals , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Spondylarthritis/blood , Spondylarthritis/epidemiology , Spondylarthritis/therapy
11.
J Clin Invest ; 130(4): 1863-1878, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32149730

ABSTRACT

Spondyloarthritis (SpA) represents a family of inflammatory diseases of the spine and peripheral joints. Ankylosing spondylitis (AS) is the prototypic form of SpA in which progressive disease can lead to fusion of the spine. Therapeutically, knowledge of type 3 immunity has translated into the development of IL-23- and IL-17A-blocking antibodies for the treatment of SpA. Despite being able to provide symptomatic control, the current biologics do not prevent the fusion of joints in AS patients. Thus, there is an unmet need for disease-modifying drugs. Genetic studies have linked the Janus kinase TYK2 to AS. TYK2 is a mediator of type 3 immunity through intracellular signaling of IL-23. Here, we describe and characterize a potentially novel small-molecule inhibitor of TYK2 that blocked IL-23 signaling in vitro and inhibited disease progression in animal models of SpA. The effect of the inhibitor appears to be TYK2 specific, using TYK2-inactive mice, which further revealed a duality in the induction of IL-17A and IL-22 by IL-23. Specifically, IL-22 production was TYK2/JAK2/STAT3 dependent, while IL-17A was mostly JAK2 dependent. Finally, we examined the effects of AS-associated TYK2 SNPs on TYK2 expression and function and correlated them with AS disease progression. This work provides evidence that TYK2 inhibitors have great potential as an orally delivered therapeutic for SpA.


Subject(s)
Polymorphism, Single Nucleotide , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Spondylarthritis , TYK2 Kinase , Animals , Humans , Interleukins/genetics , Interleukins/immunology , Mice , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Spondylarthritis/drug therapy , Spondylarthritis/genetics , Spondylarthritis/immunology , Spondylarthritis/pathology , TYK2 Kinase/antagonists & inhibitors , TYK2 Kinase/genetics , TYK2 Kinase/immunology
12.
Nat Rev Rheumatol ; 16(4): 193-207, 2020 04.
Article in English | MEDLINE | ID: mdl-32080619

ABSTRACT

Mechanical loading is an important factor in musculoskeletal health and disease. Tendons and ligaments require physiological levels of mechanical loading to develop and maintain their tissue architecture, a process that is achieved at the cellular level through mechanotransduction-mediated fine tuning of the extracellular matrix by tendon and ligament stromal cells. Pathological levels of force represent a biological (mechanical) stress that elicits an immune system-mediated tissue repair pathway in tendons and ligaments. The biomechanics and mechanobiology of tendons and ligaments form the basis for understanding how such tissues sense and respond to mechanical force, and the anatomical extent of several mechanical stress-related disorders in tendons and ligaments overlaps with that of chronic inflammatory arthritis in joints. The role of mechanical stress in 'overuse' injuries, such as tendinopathy, has long been known, but mechanical stress is now also emerging as a possible trigger for some forms of chronic inflammatory arthritis, including spondyloarthritis and rheumatoid arthritis. Thus, seemingly diverse diseases of the musculoskeletal system might have similar mechanisms of immunopathogenesis owing to conserved responses to mechanical stress.


Subject(s)
Arthritis/physiopathology , Ligaments, Articular/physiopathology , Tendons/physiopathology , Animals , Biomechanical Phenomena , Humans , Stress, Mechanical
13.
Arthritis Rheumatol ; 72(3): 428-434, 2020 03.
Article in English | MEDLINE | ID: mdl-31599089

ABSTRACT

OBJECTIVE: Ankylosing spondylitis (AS) is an inflammatory arthritis in which men have a higher risk of developing progressive axial disease than women. Transcriptomic studies have shown reduced expression of cytotoxic cell genes in the blood of AS patients. HLA-B27 contributes the greatest risk for AS, suggesting a role for CD8+ T cells. This study was undertaken to profile AS patient cytotoxic cells with the hypothesis that an alteration in CD8+ T cells might explain the aberrant cytotoxic profile observed in patients. METHODS: Whole blood was examined for GZM and PRF1 gene expression by quantitative polymerase chain reaction. Serum and synovial fluid (SF) were examined for granzyme and perforin 1 expression by bead array, and blood and SF mononuclear cells were examined for granzyme and perforin 1 expression by fluorescence-activated cell sorting (FACS). RESULTS: GZM and PRF1 gene expression were both reduced in AS patients compared to healthy controls, especially in men. Perforin 1, but not granzyme, protein levels were reduced in AS patient serum. Granzymes were elevated in AS SF, but not in rheumatoid arthritis or osteoarthritis SF. FACS revealed a reduction in granzyme-positive and perforin 1-positive lymphocytes, but not an intrinsic defect in CD8+ T cell granzyme or perforin 1 production. CD8+ T cell frequency was reduced in the blood and increased in the SF of AS patients. CONCLUSION: Our findings indicate that AS patients have an altered cytotoxic T cell profile. These data suggest that CD8+ T cells with a cytotoxic phenotype are recruited to the joints, where they exhibit an activated phenotype. Thus, a central role for CD8+ T cells in AS may have been overlooked and deserves further study.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic/genetics , Granzymes/immunology , Perforin/immunology , Spondylitis, Ankylosing/immunology , Adult , Female , Humans , Male , Middle Aged , Spondylitis, Ankylosing/genetics
14.
Ann Rheum Dis ; 78(11): 1566-1575, 2019 11.
Article in English | MEDLINE | ID: mdl-31471299

ABSTRACT

OBJECTIVES: Current evidence suggests that immune events in the gut may impact joint inflammation in ankylosing spondylitis (AS) but the expression of gut-related trafficking molecules in the inflammed joint is poorly characterised. We aimed to (1) assess differential expression patterns of trafficking molecules between patients and controls, (2) generate joint-specific cellular signatures and (3) obtain transcriptomic profiles of noteworthy cell subpopulations. METHODS: Male subjects under 40 years of age fulfilling the mNY criteria were recruited. The following cells were surface stained using a 36-marker mass cytometry antibody panel: (1) peripheral blood mononuclear cells from AS patients, and healthy controls; (2) synovial fluid mononuclear cells from AS and rheumatoid arthritis (RA) patients. Additionally, RNA-seq was performed on CD8+ T cell subpopulations from the synovial fluid (SF). RESULTS: Mature CD8+ T cells were enriched in AS SF, with a distinct pattern of integrin expression (ß7, CD103, CD29 and CD49a). RNA-seq analysis of SF-derived CD103+CD49a+CD8+ T cells revealed elevated TNFAIP3, GZMB, PRF1 and IL-10. CONCLUSIONS: We have identified a novel integrin-expressing mature CD8+ T cell population (CD49a+CD103+ß7+CD29+) that appears to be more prevalent in AS SF than RA SF. These cells seem to possess dual cytotoxic and regulatory profiles which may play a role in AS pathogenesis.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Integrins/metabolism , Spondylarthritis/immunology , Synovial Fluid/immunology , Transcriptome/immunology , Adult , Arthritis, Rheumatoid/immunology , Humans , Leukocytes, Mononuclear , Male
15.
Curr Opin Rheumatol ; 31(1): 62-69, 2019 01.
Article in English | MEDLINE | ID: mdl-30407224

ABSTRACT

PURPOSE OF REVIEW: This article aims to review recent literature linking epithelial barrier inflammation and arthritis in spondyloarthritis (SpA), with a critical view on how they are bound by genetic, immunological and environmental ties. RECENT FINDINGS: The epithelia-joint axis has become an intense area of both basic and clinical SpA research. The penultimate goal is to understand the immunopathologic links between epithelial inflammation and arthritis in SpA. Inflammatory bowel disease (IBD) and psoriasis (PsO) have strong links to SpA at several levels. Clinically, there is a strong association of IBD, PsO and SpA. Genetically, there are many shared risk factors; however, there are also distinct differences in the genetics of the respective diseases. Immunologically, type 3 immunity, especially interleukin (IL)-17 and IL-23 dysregulation, has been shown to play a central role in IBD, PsO and SpA. Environmentally, a microbial dysbiosis has been noted in each of these diseases, but whether the microbial signature is similar between diseases is not clear, nor is the effect of dysbiosis on the immune response known. SUMMARY: It will be crucial to determine whether the relationship between epithelia inflammation and SpA is truly causal for both the understanding of pathogenesis and for future treatment strategies.


Subject(s)
Inflammatory Bowel Diseases/immunology , Psoriasis/immunology , Skin/immunology , Spondylarthritis/immunology , Gastrointestinal Microbiome , Humans , Inflammation/immunology , Inflammation/microbiology , Inflammatory Bowel Diseases/microbiology , Interleukin-17 , Psoriasis/microbiology , Skin/microbiology , Spondylarthritis/microbiology
16.
Arthritis Rheumatol ; 69(9): 1796-1806, 2017 09.
Article in English | MEDLINE | ID: mdl-28597514

ABSTRACT

OBJECTIVE: To investigate the role of macrophage migration inhibitory factor (MIF) in the pathogenesis of ankylosing spondylitis (AS). METHODS: Patients who met the modified New York criteria for AS were recruited for the study. Healthy volunteers, rheumatoid arthritis patients, and osteoarthritis patients were included as controls. Based on the annual rate of increase in modified Stoke AS Spine Score (mSASSS), AS patients were classified as progressors or nonprogressors. MIF levels in serum and synovial fluid were quantitated by enzyme-linked immunosorbent assay. Predictors of AS progression were evaluated using logistic regression analysis. Immunohistochemical analysis of ileal tissue was performed to identify MIF-producing cells. Flow cytometry was used to identify MIF-producing subsets, expression patterns of the MIF receptor (CD74), and MIF-induced tumor necrosis factor (TNF) production in the peripheral blood. MIF-induced mineralization of osteoblast cells (SaOS-2) was analyzed by alizarin red S staining, and Western blotting was used to quantify active ß-catenin levels. RESULTS: Baseline serum MIF levels were significantly elevated in AS patients compared to healthy controls and were found to independently predict AS progression. MIF levels were higher in the synovial fluid of AS patients, and MIF-producing macrophages and Paneth cells were enriched in their gut. MIF induced TNF production in monocytes, activated ß-catenin in osteoblasts, and promoted the mineralization of osteoblasts. CONCLUSION: Our findings indicate an unexplored pathogenic role of MIF in AS and a link between inflammation and new bone formation.


Subject(s)
Disease Progression , Intramolecular Oxidoreductases/analysis , Macrophage Migration-Inhibitory Factors/analysis , Spondylitis, Ankylosing/immunology , Spondylitis, Ankylosing/pathology , Adult , Antigens, Differentiation, B-Lymphocyte/blood , Calcification, Physiologic , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Female , Histocompatibility Antigens Class II/blood , Humans , Logistic Models , Macrophages/metabolism , Male , Middle Aged , Monocytes/metabolism , Osteoblasts/pathology , Paneth Cells/metabolism , Predictive Value of Tests , Severity of Illness Index , Spine/immunology , Spine/pathology , Spondylitis, Ankylosing/blood , Synovial Fluid/chemistry , Tumor Necrosis Factor-alpha/blood
17.
Nat Rev Rheumatol ; 13(7): 410-420, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28615730

ABSTRACT

Clinicians have commonly differentiated chronic back pain into two broad subsets: namely, non-inflammatory (or mechanical) back pain and inflammatory back pain. As the terminology suggests, the latter category, in which ankylosing spondylitis (AS) is prominent, presupposes a close link between pain and inflammation. Advances in research into the genetics and immunology of AS have improved our understanding of the inflammatory processes involved in this disease, and have led to the development of potent anti-inflammatory biologic therapeutic agents. However, evidence from clinical trials and from biomarker and imaging studies in patients with AS indicate that pain and inflammation are not always correlated. Thus, the assumption that pain in AS is a reliable surrogate marker for inflammation might be an over-simplification. This Review provides an overview of current concepts relating to neuro-immune interactions in AS and summarizes research that reveals an increasingly complex interplay between the activation of the immune system and pain pathways in the nervous system. The different types of pain experienced by patients with AS, insights from brain imaging studies, neurological mechanisms of pain, sex bias in pain and how the immune system can modify pain in patients with AS are also discussed.


Subject(s)
Back Pain/etiology , Spondylitis, Ankylosing/complications , Back Pain/drug therapy , Back Pain/immunology , Brain/diagnostic imaging , Humans , Pain Management/methods , Spondylitis, Ankylosing/immunology
19.
Nat Rev Rheumatol ; 13(6): 359-367, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28446810

ABSTRACT

Over the past 5 years, advances in high-throughput techniques and studies involving large cohorts of patients have led to considerable advances in the identification of novel genetic associations and immune pathways involved in ankylosing spondylitis (AS). These discoveries include genes encoding cytokine receptors, transcription factors, signalling molecules and transport proteins. Although progress has been made in understanding the functions and potential pathogenic roles of some of these molecules, much work remains to be done to comprehend their complex interactions and therapeutic potential in AS. In this Review, we outline the current knowledge of AS pathogenesis, including genetic risk associations, HLA-B27-mediated pathology, perturbations in antigen-presentation pathways and the contribution of the type 3 immune response.


Subject(s)
Spondylitis, Ankylosing/etiology , Antigen Presentation/physiology , Humans , Interleukin-17/immunology , Spondylitis, Ankylosing/genetics , Spondylitis, Ankylosing/immunology
20.
J Immunol ; 198(10): 3949-3962, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28373584

ABSTRACT

Invariant NKT (iNKT) cells are innate lymphocytes that respond to glycolipids presented by the MHC class Ib molecule CD1d and are rapidly activated to produce large quantities of cytokines and chemokines. iNKT cell development uniquely depends on interactions between double-positive thymocytes that provide key homotypic interactions between signaling lymphocyte activation molecule (SLAM) family members. However, the role of SLAM receptors in the differentiation of iNKT cell effector subsets and activation has not been explored. In this article, we show that C57BL/6 mice containing the New Zealand Black Slam locus have profound alterations in Ly108, CD150, and Ly9 expression that is associated with iNKT cell hyporesponsiveness. This loss of function was only apparent when dendritic cells and iNKT cells had a loss of SLAM receptor expression. Using small interfering RNA knockdowns and peptide-blocking strategies, we demonstrated that trans-Ly108 interactions between dendritic cells and iNKT cells are critical for robust activation. LY108 costimulation similarly increased human iNKT cell activation. Thus, in addition to its established role in iNKT cell ontogeny, Ly108 regulates iNKT cell function in mice and humans.


Subject(s)
Antigens, Ly/metabolism , Dendritic Cells/metabolism , Lymphocyte Activation , Natural Killer T-Cells/immunology , Signaling Lymphocytic Activation Molecule Family Member 1/metabolism , Signaling Lymphocytic Activation Molecule Family/metabolism , Animals , Antigens, CD1d/immunology , Antigens, Ly/genetics , Antigens, Ly/immunology , Cell Differentiation , Cytokines/biosynthesis , Cytokines/immunology , Dendritic Cells/immunology , Gene Expression Regulation , Humans , Mice , Mice, Inbred C57BL , Natural Killer T-Cells/metabolism , RNA, Small Interfering , Signaling Lymphocytic Activation Molecule Family/deficiency , Signaling Lymphocytic Activation Molecule Family/genetics , Signaling Lymphocytic Activation Molecule Family/immunology , Signaling Lymphocytic Activation Molecule Family Member 1/genetics , Signaling Lymphocytic Activation Molecule Family Member 1/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...