Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Arthritis Rheumatol ; 75(7): 1152-1165, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36657110

RESUMEN

OBJECTIVE: Spondyloarthritis (SpA) is characterized by pathologic osteogenesis, inflammation, and extensive angiogenesis in axial and peripheral tissues. Current therapies effectively target inflammation, but these therapies lack efficacy in preventing pathologic osteogenesis. Transgenic mice overexpressing transmembrane tumor necrosis factor (tmTNF-Tg mice) exhibit SpA-like features. We hypothesized that type H blood vessels, which are implicated in osteogenesis, are increased and contribute to pathology in this experimental SpA model. METHODS: We analyzed ankles, femora, and vertebrae of tmTNF-Tg mice and nontransgenic littermates and tmTNF-Tg mice on either a TNF receptor type I (TNFRI)-deficient or TNF receptor type II (TNFRII)-deficient background for osteogenesis, angiogenesis, and inflammation using advanced imaging technologies at various stages of disease. RESULTS: Compared to nontransgenic littermates, tmTNF-Tg mice exhibited an increase in vertebral type H vessels and osteoprogenitor cells in subchondral bone. These features of increased angiogenesis and osteogenesis were already present before onset of clinical disease symptoms. Type H vessels and osteoprogenitor cells were in close proximity to inflammatory lesions and ectopic lymphoid structures. The tmTNF-Tg mice also showed perivertebral ectopic type H vessels and osteogenesis, an increased number of vertebral transcortical vessels, and enhanced entheseal angiogenesis. In tmTNF-Tg mice crossed on a TNFRI- or TNFRII-deficient background, no clear reduction in type H vessels was shown, suggesting that type H vessel formation is not exclusively mediated via TNFRI or TNFRII. CONCLUSION: The contribution of type H vessels to pathologic osteogenesis in experimental SpA advances our knowledge of the pathophysiology of this disease and may also provide a novel opportunity for targeted intervention.


Asunto(s)
Osteogénesis , Espondiloartritis , Ratones , Animales , Inflamación , Espondiloartritis/tratamiento farmacológico , Ratones Transgénicos , Factor de Necrosis Tumoral alfa
2.
Arthritis Care Res (Hoboken) ; 74(12): 2076-2084, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-34219406

RESUMEN

OBJECTIVE: As first-degree relatives (FDRs) of HLA-B27-positive patients with axial spondyloarthritis (SpA) have an increased risk of developing axial SpA, the objectives were 1) to evaluate the presence of highly specific imaging features as well as clinical signs of SpA at baseline and after 1 year of follow-up, and 2) to describe the evolution toward clinical disease within 1 year of follow-up in a cohort of seemingly healthy FDRs of HLA-B27-positive axial SpA patients. METHODS: The Pre-SpA cohort is a 5-year prospective inception cohort of seemingly healthy FDRs of HLA-B27-positive axial SpA patients. Clinical and imaging features were collected and recorded. RESULTS: At baseline, 19% of the FDRs reported inflammatory back pain, 32% current arthralgia, 3% arthritis (ever), 5% enthesitis (ever), and 1% dactylitis (ever), and 3% had an extraarticular manifestation. C-reactive protein level was elevated in 16%, and erythrocyte sedimentation rate was elevated in 7%. On magnetic resonance imaging (MRI) views of sacroiliac joints, 10% had a Spondyloarthritis Research Consortium of Canada score of ≥2, 4% had a score of ≥5, and 4% had deep lesions. In total, 1% fulfilled the modified New York criteria for radiographic sacroiliitis. Clinical, MRI, and acute phase findings were equally distributed between HLA-B27-positive and -negative FDRs. After 1 year of follow-up, clinical parameters did not change on the group level, but 6% of the FDRs were clinically diagnosed with axial SpA, of whom 86% were HLA-B27-positive. CONCLUSION: Features associated with SpA or imaging abnormalities were found in up to 32% of seemingly healthy FDRs, with an equal distribution between HLA-B27-positive and -negative FDRs. Progression to clinical axial SpA within 1 year of follow-up was mainly observed in HLA-B27-positive FDRs.


Asunto(s)
Antígeno HLA-B27 , Espondiloartritis , Humanos , Antígeno HLA-B27/genética , Estudios Prospectivos , Dolor de Espalda/diagnóstico , Espondiloartritis/diagnóstico por imagen , Espondiloartritis/genética , Imagen por Resonancia Magnética/métodos , Inflamación/complicaciones
3.
Int J Mol Sci ; 22(21)2021 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-34769069

RESUMEN

C-reactive protein (CRP) is an acute-phase protein in humans that is produced in high quantities by the liver upon infection and under inflammatory conditions. Although CRP is commonly used as a marker of inflammation, CRP can also directly contribute to inflammation by eliciting pro-inflammatory cytokine production by immune cells. Since CRP is highly elevated in serum under inflammatory conditions, we have studied the CRP-induced cytokine profile of human monocytes, one of the main innate immune cell populations in blood. We identified that CRP is relatively unique in its capacity to induce production of the pro-inflammatory cytokine IL-23, which was in stark contrast to a wide panel of pattern recognition receptor (PRR) ligands. We show that CRP-induced IL-23 production was mediated at the level of gene transcription, since CRP particularly promoted gene transcription of IL23A (encoding IL-23p19) instead of IL12A (encoding IL-12p35), while PRR ligands induce the opposite response. Interestingly, when CRP stimulation was combined with PRR ligand stimulation, as for example, occurs in the context of sepsis, IL-23 production by monocytes was strongly reduced. Combined, these data identify CRP as a unique individual ligand to induce IL-23 production by monocytes, which may contribute to shaping systemic immune responses under inflammatory conditions.


Asunto(s)
Proteína C-Reactiva/metabolismo , Subunidad p19 de la Interleucina-23/metabolismo , Monocitos/metabolismo , Células Cultivadas , Humanos , Subunidad p19 de la Interleucina-23/genética , ARN Mensajero/genética , Activación Transcripcional
4.
J Immunol ; 207(9): 2337-2346, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34561228

RESUMEN

TNF is important in immune-mediated inflammatory diseases, including spondyloarthritis (SpA). Transgenic (tg) mice overexpressing transmembrane TNF (tmTNF) develop features resembling human SpA. Furthermore, both tmTNF tg mice and SpA patients develop ectopic lymphoid aggregates, but it is unclear whether these contribute to pathology. Therefore, we characterized the lymphoid aggregates in detail and studied potential alterations in the B and T cell lineage in tmTNF tg mice. Lymphoid aggregates developed in bone marrow (BM) of vertebrae and near the ankle joints prior to the first SpA features and displayed characteristics of ectopic lymphoid structures (ELS) including presence of B cells, T cells, germinal centers, and high endothelial venules. Detailed flow cytometric analyses demonstrated more germinal center B cells with increased CD80 and CD86 expression, along with significantly more T follicular helper, T follicular regulatory, and T regulatory cells in tmTNF tg BM compared with non-tg controls. Furthermore, tmTNF tg mice exhibited increased IgA serum levels and significantly more IgA+ plasma cells in the BM, whereas IgA+ plasma cells in the gut were not significantly increased. In tmTNF tg × TNF-RI-/- mice, ELS were absent, consistent with reduced disease symptoms, whereas in tmTNF tg × TNF-RII-/- mice, ELS and clinical symptoms were still present. Collectively, these data show that tmTNF overexpression in mice results in osteitis and ELS formation in BM, which may account for the increased serum IgA levels that are also observed in human SpA. These effects are mainly dependent on TNF-RI signaling and may underlie important aspects of SpA pathology.


Asunto(s)
Linfocitos B/inmunología , Médula Ósea/metabolismo , Centro Germinal/inmunología , Proteínas de la Membrana/metabolismo , Osteítis/inmunología , Espondilitis Anquilosante/inmunología , Linfocitos T/inmunología , Estructuras Linfoides Terciarias/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Médula Ósea/patología , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Inmunoglobulina A/metabolismo , Proteínas de la Membrana/genética , Ratones , Transducción de Señal , Factor de Necrosis Tumoral alfa/genética
5.
Front Immunol ; 12: 611656, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33746955

RESUMEN

Background: Psoriatic arthritis (PsA) is a chronic inflammatory joint disease within the spondyloarthritis spectrum. IL-12p40/IL-23p40 blockade reduces PsA disease activity, but its impact on synovial inflammation remains unclear. Objectives: To investigate the cellular and molecular pathways affected by IL-12p40/IL-23p40 blockade with ustekinumab in the synovium of PsA patients. Methods: Eleven PsA patients with at least one inflamed knee or ankle joint were included in a 24-week single-center open-label study and received ustekinumab 45 mg/sc according to standard care at week 0, 4, and 16. Besides clinical outcomes, synovial tissue (ST) samples were obtained by needle arthroscopy from an inflamed knee or ankle joint at baseline, week 12 and 24 and analyzed by immunohistochemistry, RNA-sequencing and real-time quantitative polymerase chain reaction (qPCR). Results: We obtained paired baseline and week 12, and paired baseline, week 12 and 24 ST samples from nine and six patients, respectively. Eight patients completed 24 weeks of clinical follow-up. At 12 weeks 6/11 patients met ACR20, 2/11 met ACR50 and 1/11 met ACR70 improvement criteria, at 24 weeks this was 3/8, 2/8 and 1/8 patients, respectively. Clinical and serological markers improved significantly. No serious adverse events occurred. We observed numerical decreases of all infiltrating cell subtypes at week 12, reaching statistical significance for CD68+ sublining macrophages. For some cell types this was even more pronounced at week 24, but clearly synovial inflammation was incompletely resolved. IL-17A and F, TNF, IL-6, IL-8, and IL-12p40 were not significantly downregulated in qPCR analysis of W12 total biopsies, only MMP3 and IL-23p19 were significantly decreased. RNA-seq analysis revealed 178 significantly differentially expressed genes between baseline and 12 weeks (FDR 0.1). Gene Ontology and KEGG terms enrichment analyses identified overrepresentation of biological processes as response to reactive oxygen species, chemotaxis, migration and angiogenesis as well as MAPK-ERK and PI3K-Akt signaling pathways among the downregulated genes and of Wnt signaling pathway among the upregulated genes. Furthermore, ACR20 responders and non-responders differed strikingly in gene expression profiles in a post-hoc exploratory analysis. Conclusions: Ustekinumab suppresses PsA synovial inflammation through modulation of multiple signal transduction pathways, including MAPK-ERK, Wnt and potentially PI3K-Akt signaling rather than by directly impacting the IL-17 pathway.


Asunto(s)
Subunidad p40 de la Interleucina-12/antagonistas & inhibidores , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sinovitis/metabolismo , Sinovitis/patología , Ustekinumab/farmacología , Vía de Señalización Wnt/efectos de los fármacos , Agammaglobulinemia Tirosina Quinasa , Antirreumáticos/farmacología , Antirreumáticos/uso terapéutico , Artritis Psoriásica/tratamiento farmacológico , Artritis Psoriásica/etiología , Artritis Psoriásica/metabolismo , Artritis Psoriásica/patología , Biomarcadores , Biología Computacional/métodos , Citocinas/metabolismo , Ontología de Genes , Humanos , Inmunohistoquímica , Fosfatidilinositol 3-Quinasas/metabolismo , Índice de Severidad de la Enfermedad , Sinovitis/tratamiento farmacológico , Sinovitis/etiología , Transcriptoma , Ustekinumab/uso terapéutico
6.
Sci Rep ; 10(1): 21094, 2020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-33273664

RESUMEN

Spondyloarthritis (SpA) is characterized by inflammation and new bone formation. The exact pathophysiology underlying these processes remains elusive. We propose that the extensive neoangiogenesis in SpA could play a role both in sustaining/enhancing inflammation and in new bone formation. While ample data is available on effects of anti-TNF on angiogenesis, effects of IL-17A blockade on serum markers are largely unknown. We aimed to assess the impact of secukinumab (anti-IL-17A) on synovial neoangiogenesis in peripheral SpA, and how this related to changes in inflammatory and tissue remodeling biomarkers. Serum samples from 20 active peripheral SpA patients included in a 12 week open-label trial with secukinumab were analyzed for several markers of angiogenesis and tissue remodeling. Synovial biopsies taken before and after treatment were stained for vascular markers. Serum levels of MMP-3, osteopontin, IL-6 (all P < 0.001), IL-31, S100A8, S100A9, Vascular Endothelial Growth Factor A (VEGF-A), IL-33, TNF-α (all P < 0.05) decreased significantly upon anti-IL17A treatment. Secukinumab treatment resulted in a decrease in the number of synovial high endothelial venules and lymphoid aggregate score. These results indicate that anti-IL-17A not only diminishes inflammation, but also impacts angiogenesis and tissue remodeling/new bone formation. This may have important implications for disease progression and/or structural damage.


Asunto(s)
Antiinflamatorios/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Neovascularización Fisiológica , Espondilitis Anquilosante/tratamiento farmacológico , Biomarcadores/sangre , Interleucinas/sangre , Articulaciones/irrigación sanguínea , Articulaciones/efectos de los fármacos , Metaloproteinasa 3 de la Matriz/sangre , Osteopontina/sangre , Proteínas S100/sangre , Espondilitis Anquilosante/sangre , Factor A de Crecimiento Endotelial Vascular/sangre , Vénulas/efectos de los fármacos , Vénulas/fisiología
7.
J Immunol ; 205(12): 3400-3407, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33188071

RESUMEN

IgG Abs are crucial for various immune functions, including neutralization, phagocytosis, and Ab-dependent cellular cytotoxicity. In this study, we identified another function of IgG by showing that IgG immune complexes elicit distinct cytokine profiles by human myeloid immune cells, which are dependent on FcγR activation by the different IgG subclasses. Using monoclonal IgG subclasses with identical Ag specificity, our data demonstrate that the production of Th17-inducing cytokines, such as TNF, IL-1ß, and IL-23, is particularly dependent on IgG2, whereas type I IFN responses are controlled by IgG3, and IgG1 is able to regulate both. In addition, we identified that subclass-specific cytokine production is orchestrated at the posttranscriptional level through distinct glycolytic reprogramming of human myeloid immune cells. Combined, these data identify that IgG subclasses provide pathogen- and cell type-specific immunity through differential metabolic reprogramming by FcγRs. These findings may be relevant for future design of Ab-related therapies in the context of infectious diseases, chronic inflammation, and cancer.


Asunto(s)
Citocinas/inmunología , Inmunoglobulina G/inmunología , Células Mieloides/inmunología , Receptores de IgG/inmunología , Humanos , Células Mieloides/citología
8.
J Exp Med ; 217(10)2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32662821

RESUMEN

TNF plays a key role in immune-mediated inflammatory diseases including rheumatoid arthritis (RA) and spondyloarthritis (SpA). It remains incompletely understood how TNF can lead to different disease phenotypes such as destructive peripheral polysynovitis in RA versus axial and peripheral osteoproliferative inflammation in SpA. We observed a marked increase of transmembrane (tm) versus soluble (s) TNF in SpA versus RA together with a decrease in the enzymatic activity of ADAM17. In contrast with the destructive polysynovitis observed in classical TNF overexpression models, mice overexpressing tmTNF developed axial and peripheral joint disease with synovitis, enthesitis, and osteitis. Histological and radiological assessment evidenced marked endochondral new bone formation leading to joint ankylosis over time. SpA-like inflammation, but not osteoproliferation, was dependent on TNF-receptor I and mediated by stromal tmTNF overexpression. Collectively, these data indicate that TNF can drive distinct inflammatory pathologies. We propose that tmTNF is responsible for the key pathological features of SpA.


Asunto(s)
Artritis/metabolismo , Osteogénesis , Espondiloartritis/metabolismo , Factor de Necrosis Tumoral alfa/fisiología , Proteína ADAM17/metabolismo , Adulto , Animales , Artritis/etiología , Modelos Animales de Enfermedad , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Articulaciones/metabolismo , Masculino , Ratones , Receptores del Factor de Necrosis Tumoral/metabolismo , Espondiloartritis/etiología , Sinovitis/etiología , Sinovitis/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
9.
Arthritis Rheumatol ; 72(8): 1303-1313, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32243724

RESUMEN

OBJECTIVE: Interleukin-17A (IL-17A) and tumor necrosis factor (TNF) contribute to the pathogenesis of psoriatic arthritis (PsA). However, their functional relationship in PsA synovitis has not been fully elucidated. Additionally, although CD8+ T cells in PsA have been recognized via flow cytometry as a source of IL-17A production, it is not clear whether CD8+ T cells secrete IL-17A under more physiologically relevant conditions in the context from PsA synovitis. This study was undertaken to clarify the roles of IL-17A and TNF in the synovial fluid (SF) from patients with PsA and investigate the impact of CD8+ T cells on IL-17A production. METHODS: IL-17A+ T cells were identified by flow cytometry in SF samples from 20 patients with active PsA, blood samples from 22 treatment-naive patients with PsA, and blood samples from 22 healthy donors. IL-17A+ T cells were sorted from 12 PsA SF samples and stimulated using anti-CD3/anti-CD28 or phorbol myristate acetate (PMA) and ionomycin ex vivo, alone (n = 3) or together with autologous monocytes (n = 3) or PsA fibroblast-like synoviocytes (FLS) (n = 5-6). To evaluate the differential allogeneic effects of neutralizing IL-17A and TNF, SF CD4+ T cells and PsA FLS cocultures were also used (n = 5-6). RESULTS: Flow cytometry analyses of SF samples from patients with PsA showed IL-17A positivity for CD4+ and CD8+ T cells (IL-17A, median 0.71% [interquartile range 0.35-1.50%] in CD4+ cells; median 0.44% [interquartile range 0.17-1.86%] in CD8+ T cells). However, only CD4+ T cells secreted IL-17A after anti-CD3/anti-CD28 activation, when cultured alone and in cocultures with PsA monocytes or PsA FLS (each P < 0.05). Remarkably, CD8+ T cells only secreted IL-17A after 4- or 72-hour stimulation with PMA/ionomycin. Anti-IL-17A and anti-TNF treatments both inhibited PsA synovitis ex vivo. Neutralizing IL-17A strongly inhibited IL-6 (P < 0.05) and IL-1ß (P < 0.01), while anti-TNF treatment was more potent in reducing matrix metalloproteinase 3 (MMP-3) (P < 0.05) and MMP-13. CONCLUSION: CD8+ T cells, in contrast to CD4+ T cells, in SF specimens obtained from PsA patients did not secrete IL-17A following T cell receptor activation. Overlapping, but distinct, effects at the level of inflammatory cytokines and MMPs were found after neutralizing IL-17A or TNF ex vivo in a human model of PsA synovitis.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD8-positivos/efectos de los fármacos , Mediadores de Inflamación/metabolismo , Interleucina-17/biosíntesis , Receptores de Antígenos de Linfocitos T/administración & dosificación , Factor de Necrosis Tumoral alfa/metabolismo , Adulto , Artritis Psoriásica/tratamiento farmacológico , Artritis Psoriásica/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Técnicas de Cultivo de Célula , Femenino , Citometría de Flujo , Humanos , Ionomicina/farmacología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Masculino , Receptores de Antígenos de Linfocitos T/inmunología , Líquido Sinovial , Sinoviocitos/efectos de los fármacos , Sinoviocitos/inmunología , Sinovitis/tratamiento farmacológico , Sinovitis/inmunología , Acetato de Tetradecanoilforbol/farmacología
10.
J Rheumatol ; 47(11): 1606-1613, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31941804

RESUMEN

OBJECTIVE: Targeting the interleukin 17 (IL-17) axis is efficacious in psoriasis and spondyloarthritis (SpA), but not in rheumatoid arthritis (RA). We investigated potential differences in tissue expression and function of IL-17A and IL-17F in these conditions. METHODS: mRNA expression of cytokines and their receptors was assessed by quantitative PCR in psoriasis skin samples, in SpA and RA synovial tissue (ST) samples and in fibroblast-like synoviocytes (FLS). Cytokines were measured in synovial fluid (SF) and FLS supernatants by ELISA. FLS were stimulated with IL-17A or IL-17F cytokines supplemented with tumor necrosis factor (TNF), or with pooled SF from patients with SpA or RA. RESULTS: Levels of IL-17A (P = 0.031) and IL-17F (P = 0.017) mRNA were lower in psoriatic arthritis ST compared to paired psoriasis skin samples. The level of IL-17A mRNA was 2.7-fold lower than that of IL-17F in skin (P = 0.0078), but 17.3-fold higher in ST (P < 0.0001). In SF, the level of IL-17A protein was 37.4-fold higher than that of IL-17F [median 292.4 (IQR 81.4-464.2) vs median 7.8 (IQR 7.7-8.7) pg/mL; P < 0.0001]. IL-17A and IL-17F mRNA and protein levels did not differ in SpA compared to RA synovitis samples, and neither were the IL-17 receptors IL-17RA and IL-17RC, or the TNF receptors TNFR1 and TNR2, differentially expressed between SpA and RA ST, nor between SpA and RA FLS. SpA and RA FLS produced similar amounts of IL-6 and IL-8 protein upon stimulation with IL-17A or IL-17F cytokines, supplemented with 1 ng/ml TNF. Pooled SpA or RA SF samples similarly enhanced the inflammatory response to IL-17A and IL-17F simulation in FLS. CONCLUSION: The IL-17A/IL-17F expression ratio is higher in SpA synovitis compared to psoriasis skin. Expression of IL-17A and IL-17F, and the functional response to these cytokines, appear to be similar in SpA and RA synovitis.


Asunto(s)
Artritis Reumatoide , Espondiloartritis , Sinoviocitos , Artritis Reumatoide/inmunología , Células Cultivadas , Humanos , Interleucina-17 , Espondiloartritis/inmunología , Membrana Sinovial
11.
Arthritis Res Ther ; 21(1): 208, 2019 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-31521192

RESUMEN

BACKGROUND: If TNF inhibitors are initiated in the early stages of psoriatic arthritis, this could potentially modulate disease and therefore allow us to discontinue the TNF inhibitor after achieving remission. OBJECTIVE: To investigate whether remission induced by tumour necrosis factor alpha inhibitor (TNFi) and methotrexate in patients with early psoriatic arthritis is sustained after withdrawal of TNFi. METHODS: Open-label extension of a recently published double-blind, randomized placebo-controlled trial. Patients with psoriatic arthritis fulfilling the CASPAR criteria and with active disease at baseline (swollen and tender joint count ≥ 3) were randomized to either golimumab and methotrexate or matched placebo and methotrexate. Patients in Disease Activity Score (DAS) remission at week 22 continued in the open-label extension on methotrexate monotherapy. The primary end point was the percentage of patients in DAS-CRP remission (DAS < 1.6) at week 50. RESULTS: Eight patients from the original placebo group and 18 patients from the original TNFi group continued in the extension phase. At week 50, 6 out of 8 (75%) patients from the original MTX (methotrexate) group versus 10 out of 18 (56%) patients from the original MTX+TNFi group were in DAS-CRP remission (p = 0.347). Considering the total study population, 6 out of 24 (25%) of the original MTX group versus 10 out of 26 (38.5%) of the original MTX+TNFi group were in DAS remission at week 50 (p = 0.308). CONCLUSIONS: Remission achieved by initial combination treatment with TNFi and methotrexate in early psoriatic arthritis is maintained on methotrexate monotherapy in approximately half of the patients. TRIAL REGISTRATION: Registered at Clinicaltrials.gov with number NCT01871649 on June 7, 2013.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Artritis Psoriásica/tratamiento farmacológico , Metotrexato/administración & dosificación , Inducción de Remisión/métodos , Inhibidores del Factor de Necrosis Tumoral/uso terapéutico , Antirreumáticos/administración & dosificación , Artritis Psoriásica/diagnóstico , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Quimioterapia Combinada , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Factores de Tiempo , Resultado del Tratamiento
12.
Front Immunol ; 10: 1368, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31258536

RESUMEN

Regulatory B cells (Bregs) are immunosuppressive cells that modulate immune responses through multiple mechanisms. The signals required for the differentiation and activation of these cells remain still poorly understood. We have already shown that overexpression of A PRoliferation-Inducing Ligand (APRIL) reduces the incidence and severity of collagen-induced arthritis (CIA) in mice. Furthermore, we have described that APRIL, but not BAFF, promoted IL-10 production and regulatory functions in human B cells. Therefore, we hypothesized that APRIL, but not BAFF, may be involved in the induction and/or activation of IL-10 producing Bregs that suppress inflammatory responses in vitro and in vivo. Here, we describe that APRIL promotes the differentiation of naïve human B cells to IL-10-producing IgA+ B cells. These APRIL-induced IgA+ B cells display a Breg phenotype and inhibit T cell and macrophage responses through IL-10 and PD-L1. Moreover, APRIL-induced IL-10 producing Bregs suppress inflammation in vivo in experimental autoimmune encephalitis (EAE) and contact hypersensitivity (CHS) models. Finally, we showed a strong correlation between APRIL and IL-10 in the inflamed synovial tissue of inflammatory arthritis patients. Collectively, these observations indicate the potential relevance of this novel APRIL-induced IgA+ Breg population for immune homeostasis and immunopathology.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Linfocitos B Reguladores/inmunología , Dermatitis por Contacto/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Inflamación/inmunología , Esclerosis Múltiple/inmunología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Animales , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Tolerancia Inmunológica , Inmunoglobulina A/metabolismo , Interleucina-10/genética , Interleucina-10/metabolismo , Ratones , Ratones Transgénicos , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
13.
Inflamm Res ; 68(9): 775-785, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31227842

RESUMEN

OBJECTIVE AND DESIGN: To determine whether ER stress affects the inhibitory pathways of the human immune system, particularly the immunosuppressive effect of IL-10 on macrophages. MATERIAL OR SUBJECTS: In vitro stimulation of human monocyte-derived macrophages. TREATMENT: Cells were stimulated with TLR ligands and IL-10, while ER stress was induced using thapsigargin or tunicamycin. METHODS: mRNA expression was determined using qPCR, while cytokine protein production was measured using ELISA. Protein expression of receptors and transcription factors was determined using flow cytometry. Student's t test was used for statistics. RESULTS: While under normal conditions IL-10 potently suppresses pro-inflammatory cytokine production by LPS-stimulated macrophages, we demonstrate that ER stress counteracts the immunosuppressive effects of IL-10, leading to increased pro-inflammatory cytokine production. We identified that ER stress directly interferes with IL-10R signaling by reducing STAT3 phosphorylation on Tyr705, which thereby inhibits the expression of SOCS3. Moreover, we show that ER stress also inhibits STAT3 activation induced by other receptors such as IL-6R. CONCLUSIONS: Combined, these data uncover a new general mechanism by which ER stress promotes inflammation. Considering its potential involvement in the pathogenesis of diseases such as Crohn's disease and spondyloarthritis, targeting of this mechanism may provide new opportunities to counteract inflammation.


Asunto(s)
Estrés del Retículo Endoplásmico , Interleucina-10/farmacología , Macrófagos/citología , Factor de Transcripción STAT3/metabolismo , Humanos , Terapia de Inmunosupresión , Inflamación , Ligandos , Lipopolisacáridos/farmacología , Monocitos/citología , Fosforilación , Factor de Transcripción STAT3/antagonistas & inhibidores , Transducción de Señal , Tapsigargina/farmacología , Tunicamicina/farmacología
14.
J Immunol ; 203(1): 225-235, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31118224

RESUMEN

C-reactive protein (CRP) is an acute-phase protein produced in high quantities by the liver in response to infection and during chronic inflammatory disorders. Although CRP is known to facilitate the clearance of cell debris and bacteria by phagocytic cells, the role of CRP in additional immunological functions is less clear. This study shows that complexed CRP (phosphocholine [PC]:CRP) (formed by binding of CRP to PC moieties), but not soluble CRP, synergized with specific TLRs to posttranscriptionally amplify TNF, IL-1ß, and IL-23 production by human inflammatory macrophages. We identified FcγRI and IIa as the main receptors responsible for initiating PC:CRP-induced inflammation. In addition, we identified the underlying mechanism, which depended on signaling through kinases Syk, PI3K, and AKT2, as well as glycolytic reprogramming. These data indicate that in humans, CRP is not only a marker but also a driver of inflammation by human macrophages. Therefore, although providing host defense against bacteria, PC:CRP-induced inflammation may also exacerbate pathology in the context of disorders such as atherosclerosis.


Asunto(s)
Proteína C-Reactiva/metabolismo , Inflamación/inmunología , Hígado/fisiología , Receptores de IgG/metabolismo , Aterosclerosis/inmunología , Proteína C-Reactiva/química , Células Cultivadas , Reprogramación Celular , Citocinas/metabolismo , Glucólisis , Humanos , Mediadores de Inflamación/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilcolina/química , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Quinasa Syk/metabolismo , Receptores Toll-Like/metabolismo
15.
J Autoimmun ; 101: 131-144, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31053401

RESUMEN

During T cell-dependent (TD) germinal center (GC) responses, naïve B cells are instructed to differentiate towards GC B cells (GCBC), high-affinity long-lived plasma cells (LLPC) or memory B cells (Bmem). Alterations in the B cell-fate choice could contribute to immune dysregulation leading to the loss of self-tolerance and the initiation of autoimmune disease. Here we show that mRNA levels of the transcription regulator BOB.1 are increased in the lymph node compartment of patients with rheumatoid arthritis (RA), a prototypical autoimmune disease caused by the loss of immunological tolerance. Investigating to what extent levels of BOB.1 impact B cells during TD immune responses we found that BOB.1 has a crucial role in determining the B cell-fate decision. High BOB.1 levels promote the generation of cells with phenotypic and functional characteristics of Bmem. Mechanistically, overexpression of BOB.1 drives ABF1 and suppresses BCL6, favouring Bmem over LLPC or recycling GCBC. Low levels of BOB.1 are sufficient for LLPC but not for Bmem differentiation. Our findings demonstrate a novel role for BOB.1 in B cells during TD GC responses and suggest that its dysregulation may contribute to the pathogenesis of RA by disturbing the B cell-fate determination.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/metabolismo , Centro Germinal/inmunología , Centro Germinal/metabolismo , Memoria Inmunológica/genética , Transactivadores/genética , Animales , Biomarcadores , Línea Celular , Expresión Génica , Humanos , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Ratones , Ratones Noqueados , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Fiebre Reumática/genética , Fiebre Reumática/inmunología , Fiebre Reumática/metabolismo , Fiebre Reumática/patología , Linfocitos T/inmunología , Linfocitos T/metabolismo
16.
Front Immunol ; 10: 739, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31024565

RESUMEN

Antigen-presenting cells (APCs) such as dendritic cells (DCs) are crucial for initiation of adequate inflammatory responses, which critically depends on the cooperated engagement of different receptors. In addition to pattern recognition receptors (PRRs), Fc gamma receptors (FcγRs) have recently been identified to be important in induction of inflammation by DCs. FcγRs that recognize IgG immune complexes, which are formed upon opsonization of pathogens, induce pro-inflammatory cytokine production through cross-talk with PRRs such as Toll-like receptors (TLRs). While the physiological function of FcγR-TLR cross-talk is to provide protective immunity against invading pathogens, undesired activation of FcγR-TLR cross-talk, e.g., by autoantibodies, also plays a major role in the development of chronic inflammatory disorders such as rheumatoid arthritis (RA). Yet, the molecular mechanisms of FcγR-TLR cross-talk are still largely unknown. Here, we identified that FcγR-TLR cross-talk-induced cytokine production critically depends on activation of the transcription factor interferon regulatory factor 5 (IRF5), which results from induction of two different pathways that converge on IRF5 activation. First, TLR stimulation induced phosphorylation of TBK1/IKKε, which is required for IRF5 phosphorylation and subsequent activation. Second, FcγR stimulation induced nuclear translocation of IRF5, which is essential for gene transcription by IRF5. We identified that IRF5 activation by FcγR-TLR cross-talk amplifies pro-inflammatory cytokine production by increasing cytokine gene transcription, but also by synergistically inducing glycolytic reprogramming, which is another essential process for induction of inflammatory responses by DCs. Combined, here we identified IRF5 as a pivotal component of FcγR-TLR cross-talk in human APCs. These data may provide new potential targets to suppress chronic inflammation in autoantibody-associated diseases that are characterized by undesired or excessive FcγR-TLR cross-talk, such as RA, systemic sclerosis, and systemic lupus erythematous.


Asunto(s)
Células Dendríticas/inmunología , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Receptores de IgG/metabolismo , Receptores Toll-Like/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis , Transporte Activo de Núcleo Celular , Células Dendríticas/metabolismo , Glucólisis/inmunología , Humanos , Quinasa I-kappa B/inmunología , Quinasa I-kappa B/metabolismo , Técnicas In Vitro , Inflamación/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Modelos Inmunológicos , Monocitos/inmunología , Monocitos/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Cross-Talk/inmunología , Transcripción Genética
18.
Cell Mol Life Sci ; 76(6): 1041-1055, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30498997

RESUMEN

The prevailing concept regarding the immunological function of immunoglobulin A (IgA) is that it binds to and neutralizes pathogens to prevent infection at mucosal sites of the body. However, recently, it has become clear that in humans IgA is also able to actively contribute to the initiation of inflammation, both at mucosal and non-mucosal sites. This additional function of IgA is initiated by the formation of immune complexes, which trigger Fc alpha Receptor I (FcαRI) to synergize with various other receptors to amplify inflammatory responses. Recent findings have demonstrated that co-stimulation of FcαRI strongly affects pro-inflammatory cytokine production by various myeloid cells, including different dendritic cell subsets, macrophages, monocytes, and Kupffer cells. FcαRI-induced inflammation plays a crucial role in orchestrating human host defense against pathogens, as well as the generation of tissue-specific immunity. In addition, FcαRI-induced inflammation is suggested to be involved in the pathogenesis of various chronic inflammatory disorders, including inflammatory bowel disease, celiac disease, and rheumatoid arthritis. Combined, IgA-induced inflammation may be used to either promote inflammatory responses, e.g. in the context of cancer therapy, but may also provide new therapeutic targets to counteract chronic inflammation in the context of various chronic inflammatory disorders.


Asunto(s)
Antígenos/inmunología , Inmunoglobulina A/inmunología , Inflamación/inmunología , Membrana Mucosa/inmunología , Antígenos/metabolismo , Antígenos CD/inmunología , Antígenos CD/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Humanos , Inmunoglobulina A/metabolismo , Modelos Inmunológicos , Membrana Mucosa/metabolismo , Células Mieloides/inmunología , Células Mieloides/metabolismo , Unión Proteica , Receptores Fc/inmunología , Receptores Fc/metabolismo
19.
Arthritis Rheumatol ; 71(3): 392-402, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30260078

RESUMEN

OBJECTIVE: Clinical trials of the anti-interleukin-17A (anti-IL-17A) antibody secukinumab have demonstrated a crucial role of the cytokine IL-17A in the pathogenesis of spondyloarthritis (SpA); however, its cellular source in this condition remains a matter of controversy. Group 3 innate lymphoid cells (ILC3s) have been recently identified as potent producers of proinflammatory cytokines, including IL-17A and IL-22, in a number of different tissues. This study was undertaken to characterize the presence and composition of ILCs, and investigate whether these cells are an important source of IL-17A, in the synovial tissue (ST) of patients with SpA. METHODS: Matched ST, synovial fluid, and peripheral blood (PB) samples were obtained from SpA patients with actively inflamed knee joints. ILC subsets were characterized by flow cytometry. Gene expression analysis at the single-cell level was performed directly ex vivo and after in vitro activation. An IL-17A enzyme-linked immunospot assay was used to detect IL-17A-secreting cells. RESULTS: ILCs, and particularly NKp44+ ILC3s, were expanded in inflamed arthritic joints. Single-cell expression analysis demonstrated that ST ILCs were clearly distinguishable from ST T cells and from their PB counterparts. Expression of the Th17 signature transcripts RORC, AHR, and IL23R was detected in a large proportion of ST ILC3s. These cells were capable of inducing expression of IL22 and CSF2, but not IL17A, in response to in vitro restimulation. CONCLUSION: Our findings demonstrate that absolute and relative numbers of ILC3s are enriched in the synovial joints of patients with SpA. However, these cells are not a significant source of IL-17A in this disease.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Inmunidad Innata/inmunología , Interleucina-17/inmunología , Interleucinas/inmunología , Linfocitos/inmunología , Espondiloartritis/inmunología , Adulto , Artritis Reumatoide/inmunología , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Persona de Mediana Edad , Espondiloartritis/patología , Líquido Sinovial/inmunología , Interleucina-22
20.
Eur J Immunol ; 48(11): 1796-1809, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30184252

RESUMEN

Type I and type III interferons (IFNs) are fundamental for antiviral immunity, but prolonged expression is also detrimental to the host. Therefore, upon viral infection high levels of type I and III IFNs are followed by a strong and rapid decline. However, the mechanisms responsible for this suppression are still largely unknown. Here, we show that IgG opsonization of model viruses influenza and respiratory syncytial virus (RSV) strongly and selectively suppressed type I and III IFN production by various human antigen-presenting cells. This suppression was induced by selective inhibition of TLR, RIG-I-like receptor, and STING-dependent type I and III IFN gene transcription. Surprisingly, type I and III IFN suppression was mediated by Syk and PI3K independent inhibitory signaling via FcγRIIa, thereby identifying a novel non-canonical FcγRIIa pathway in myeloid cells. Together, these results indicate that IgG opsonization of viruses functions as a novel negative feedback mechanism in humans, which may play a role in the selective suppression of type I and III IFN responses during the late-phase of viral infections. In addition, activation of this pathway may be used as a tool to limit type I IFN-associated pathology.


Asunto(s)
Interferón Tipo I/inmunología , Interferones/inmunología , Células Mieloides/inmunología , Receptores de IgG/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Células Cultivadas , Femenino , Humanos , Inmunoglobulina G/inmunología , Ratones , Ratones Endogámicos C57BL , Virus Sincitiales Respiratorios/inmunología , Transducción de Señal/inmunología , Quinasa Syk/inmunología , Transcripción Genética/inmunología , Virosis/inmunología , Interferón lambda
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...