Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
2.
Arthritis Rheumatol ; 76(4): 531-540, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37984422

RESUMEN

OBJECTIVE: We analyzed the impact of amino acid (AA) availability on the inflammatory response in arthritis. METHODS: We stimulated rheumatoid arthritis (RA) fibroblast-like synoviocytes (FLSs) with tumor necrosis factor (TNF) in the presence or absence of proteinogenic AAs and measured their response by QuantSeq 3' messenger RNA sequencing, quantitative polymerase chain reaction, and enzyme-linked immunosorbent assay. Signal transduction events were determined by Western blot. We performed K/BxN serum transfer arthritis in mice receiving a normal and a low-protein diet and analyzed arthritis clinically and histologically. RESULTS: Deprivation of AAs decreased the expression of a specific subset of genes, including the chemokines CXCL10, CCL2, and CCL5 in TNF-stimulated FLSs. Mechanistically, the presence of AAs was required for the TNF-induced activation of an interferon regulatory factor 1 (IRF1)-STAT1 signaling circuit that drives the expression of chemotactic factors. The expression of IRF1 and the IRF1-dependent gene set in FLSs was highly correlated with the presence of inflammatory cells in human RA, emphasizing the important role of this AA-dependent pathway in inflammatory cell recruitment to the synovial tissue. Finally, we show that mice receiving a low-protein diet expressed less IRF1 in the inflamed synovium and consequently developed reduced clinical and histologic signs of arthritis. CONCLUSION: AA deprivation reduces the severity of arthritis by suppressing the expression of IRF1-STAT1-driven chemokines, which are crucial for leukocyte recruitment to the arthritic joint. Overall, our study provides novel insights into critical determinants of inflammatory arthritis and may pave the way for dietary intervention trials in RA.


Asunto(s)
Artritis Reumatoide , Sinoviocitos , Humanos , Ratones , Animales , Sinoviocitos/metabolismo , Aminoácidos/metabolismo , Artritis Reumatoide/genética , Factor de Necrosis Tumoral alfa/metabolismo , Quimiocina CXCL10/metabolismo , Aminas/metabolismo , Fibroblastos/metabolismo , Leucocitos/metabolismo , Leucocitos/patología , Células Cultivadas
3.
Ann Rheum Dis ; 82(9): 1142-1152, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37344156

RESUMEN

INTRODUCTION: Structural reorganisation of the synovium with expansion of fibroblast-like synoviocytes (FLS) and influx of immune cells is a hallmark of rheumatoid arthritis (RA). Activated FLS are increasingly recognised as a critical component driving synovial tissue remodelling by interacting with immune cells resulting in distinct synovial pathotypes of RA. METHODS: Automated high-content fluorescence microscopy of co-cultured cytokine-activated FLS and autologous peripheral CD4+ T cells from patients with RA was established to quantify cell-cell interactions. Phenotypic profiling of cytokine-treated FLS and co-cultured T cells was done by flow cytometry and RNA-Seq, which were integrated with publicly available transcriptomic data from patients with different histological synovial pathotypes. Computational prediction and knock-down experiments were performed in FLS to identify adhesion molecules for cell-cell interaction. RESULTS: Cytokine stimulation, especially with TNF-α, led to enhanced FLS-T cell interaction resulting in cell-cell contact-dependent activation, proliferation and differentiation of T cells. Signatures of cytokine-activated FLS were significantly enriched in RA synovial tissues defined as lymphoid-rich or leucocyte-rich pathotypes, with the most prominent effects for TNF-α. FLS cytokine signatures correlated with the number of infiltrating CD4+ T cells in synovial tissue of patients with RA. Ligand-receptor pair interaction analysis identified ICAM1 on FLS as an important mediator in TNF-mediated FLS-T cell interaction. Both, ICAM1 and its receptors were overexpressed in TNF-treated FLS and co-cultured T cells. Knock-down of ICAM1 in FLS resulted in reduced TNF-mediated FLS-T cell interaction. CONCLUSION: Our study highlights the role of cytokine-activated FLS in orchestrating inflammation-associated synovial pathotypes providing novel insights into disease mechanisms of RA.


Asunto(s)
Artritis Reumatoide , Sinoviocitos , Humanos , Citocinas , Factor de Necrosis Tumoral alfa/farmacología , Membrana Sinovial/patología , Sinoviocitos/patología , Fibroblastos/patología , Células Cultivadas
5.
Ann Rheum Dis ; 81(7): 1017-1022, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35304407

RESUMEN

OBJECTIVE: To assess the humoral response to messenger RNA (mRNA) vaccine of patients with systemic autoimmune rheumatic disease (SARD) and the effect of immunosuppressive medication in a matched cohort study. METHODS: Patients with SARD were enrolled and matched 1:1 for sex and age with healthy control (HC) subjects. Differences in humoral response to two doses of an mRNA vaccine in terms of seroconversion rate (SCR) and SARS-CoV-2 antibody level between the two groups and the impact of treatment within patients with SARD were assessed. RESULTS: We enrolled 82 patients with SARD and 82 matched HC. SCR after the first dose was lower among the patient group than that of HC (65% compared with 100% in HC, p<0.0001) but levelled up after the second dose (94% vs 100%). After the second dose, SCR was lower for patients on combination disease-modifying antirheumatic drug (DMARD) therapy compared with all other groups (81% compared with 95% for monotherapy, p=0.01; 100% for both no DMARD therapy and HC, both p<0.0001). In addition, antibody levels after both doses were lower in patients compared with HC. We found that vaccination response was determined primarily by the number of DMARDs and/or glucocorticoids received, with patients receiving combination therapy (dual and triple therapy) showing the poorest response. CONCLUSIONS: Patients with SARD showed a good response after the second vaccination with an mRNA vaccine. However, the choice of immunosuppressive medication has a marked effect on both SCR and overall antibody level, and the number of different immunomodulatory therapies determines vaccination response.


Asunto(s)
Antirreumáticos , COVID-19 , Enfermedades Reumáticas , Antirreumáticos/uso terapéutico , COVID-19/prevención & control , Vacunas contra la COVID-19 , Estudios de Cohortes , Humanos , Inmunosupresores/uso terapéutico , Estudios Prospectivos , Enfermedades Reumáticas/tratamiento farmacológico , SARS-CoV-2 , Vacunación , Vacunas Sintéticas , Vacunas de ARNm
6.
Rheumatology (Oxford) ; 61(11): 4535-4546, 2022 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-35258553

RESUMEN

OBJECTIVES: TNF-induced activation of fibroblast-like synoviocytes (FLS) is a critical determinant for synovial inflammation and joint destruction in RA. The detrimental role of TNF-receptor 1 (TNFR1) has thoroughly been characterized. The contributions of TNFR2, however, are largely unknown. This study was performed to delineate the role of TNFR2 in human FLS activation. METHODS: TNFR2 expression in synovial tissue samples was determined by immunohistochemistry. Expression of TNFR2 was silenced using RNAi or CRISPR/Cas9 technologies. Global transcriptional changes were determined by RNA-seq. QPCR, ELISA and immunoblotting were used to validate RNA-seq results and to uncover pathways operating downstream of TNFR2 in FLS. RESULTS: TNFR2 expression was increased in RA when compared with OA synovial tissues. In particular, RA-FLS demonstrated higher levels of TNFR2 when compared with OA-FLS. TNFR2 expression in RA-FLS correlated with RA disease activity, synovial T- and B-cell infiltration. TNF and IL1ß were identified as inflammatory mediators that upregulate TNFR2 in RA-FLS. Silencing of TNFR2 in RA-FLS markedly diminished the TNF-induced expression of inflammatory cytokines and chemokines, including CXCR3-binding chemokines and the B-cell activating factor TNFSF13B. Immunobiochemical analyses revealed that TNFR2-mediated expression of inflammatory mediators critically depends on STAT1. CONCLUSION: Our results define a critical role for TNFR2 in FLS-driven inflammation and unfold its participation in the unresolved course of synovial inflammation in RA.


Asunto(s)
Artritis Reumatoide , Receptores Tipo II del Factor de Necrosis Tumoral , Sinoviocitos , Humanos , Artritis Reumatoide/metabolismo , Células Cultivadas , Fibroblastos/metabolismo , Inflamación/metabolismo , Mediadores de Inflamación/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Membrana Sinovial/metabolismo , Sinoviocitos/metabolismo
7.
J Autoimmun ; 108: 102379, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31883829

RESUMEN

Rheumatoid Arthritis (RA) represents a chronic T cell-mediated inflammatory autoimmune disease. Studies have shown that epigenetic mechanisms contribute to the pathogenesis of RA. Histone deacetylases (HDACs) represent one important group of epigenetic regulators. However, the role of individual HDAC members for the pathogenesis of arthritis is still unknown. In this study we demonstrate that mice with a T cell-specific deletion of HDAC1 (HDAC1-cKO) are resistant to the development of Collagen-induced arthritis (CIA), whereas the antibody response to collagen type II was undisturbed, indicating an unaltered T cell-mediated B cell activation. The inflammatory cytokines IL-17 and IL-6 were significantly decreased in sera of HDAC1-cKO mice. IL-6 treated HDAC1-deficient CD4+ T cells showed an impaired upregulation of CCR6. Selective inhibition of class I HDACs with the HDAC inhibitor MS-275 under Th17-skewing conditions inhibited the upregulation of chemokine receptor 6 (CCR6) in mouse and human CD4+ T cells. Accordingly, analysis of human RNA-sequencing (RNA-seq) data and histological analysis of synovial tissue samples from human RA patients revealed the existence of CD4+CCR6+ cells with enhanced HDAC1 expression. Our data indicate a key role for HDAC1 for the pathogenesis of CIA and suggest that HDAC1 and other class I HDACs might be promising targets of selective HDAC inhibitors (HDACi) for the treatment of RA.


Asunto(s)
Artritis Reumatoide/etiología , Artritis Reumatoide/metabolismo , Susceptibilidad a Enfermedades , Histona Desacetilasa 1/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Artritis Reumatoide/patología , Biomarcadores , Colágeno/efectos adversos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Histona Desacetilasa 1/genética , Humanos , Mediadores de Inflamación/metabolismo , Ratones , Ratones Noqueados , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
8.
J Autoimmun ; 110: 102373, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31831256

RESUMEN

The mechanistic target of rapamycin (mTOR) is a master regulator of the inflammatory response in immune and non-immune cells. In immune cells mTOR regulates metabolism to fuel cell fate decision, proliferation and effector functions. In non-immune cells, such as fibroblast, it controls inflammation-associated proliferation and migration/invasion, shapes the expression of cytokines and chemokines and promotes extracellular matrix remodeling and fibrosis. Hence, mTOR plays a critical role in chronic inflammation, where a continuous feedback between stromal cells and infiltrating immune cells result in tissue remodeling and organ damage. Activation of mTOR has been implicated in a number of chronic inflammatory diseases, especially rheumatic diseases, such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), systemic sclerosis (SSc), sjögren syndrome (SS) and seronegative spondyloarthropathy (SpA). Here we review recent advances in our understanding of the mechanism of mTOR activation in inflammation, especially in rheumatic diseases. We further discuss recent findings regarding the beneficial and side effects of mTOR inhibition in rheumatic conditions.


Asunto(s)
Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/metabolismo , Autoinmunidad , Susceptibilidad a Enfermedades , Serina-Treonina Quinasas TOR/metabolismo , Enfermedades Autoinmunes/diagnóstico , Enfermedades Autoinmunes/terapia , Biomarcadores , Citocinas/metabolismo , Humanos , Terapia Molecular Dirigida , Especificidad de Órganos/genética , Especificidad de Órganos/inmunología , Fenotipo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
9.
Exp Mol Med ; 51(7): 1-11, 2019 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-31285419

RESUMEN

Rheumatoid arthritis (RA) is an autoimmune disease characterized by persistent synovial inflammation. The major drivers of synovial inflammation are cytokines and chemokines. Among these molecules, TNF activates fibroblast-like synoviocytes (FLSs), which leads to the production of inflammatory mediators. Here, we show that TNF regulates the expression of the transcription factor interferon regulatory factor 1 (IRF1) in human FLSs as well as in a TNF transgenic arthritis mouse model. Transcriptomic analyses of IRF1-deficient, TNF-stimulated FLSs define the interferon (IFN) pathway as a major target of IRF1. IRF1 expression is associated with the expression of IFNß, which leads to the activation of the JAK-STAT pathway. Blocking the JAK-STAT pathway with the Janus kinase inhibitor (JAKinib) baricitinib or tofacitinib reduces the expression of IFN-regulated genes (IRGs) in TNF-activated FLSs. Therefore, we conclude that TNF induces a distinct inflammatory cascade, in which IRGs are key elements, in FLSs. The IFN-signature might be a promising biomarker for the efficient and personalized use of new treatment strategies for RA, such as JAKinibs.


Asunto(s)
Artritis Reumatoide/inmunología , Factor 1 Regulador del Interferón/metabolismo , Interferones/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Artritis Reumatoide/metabolismo , Artritis Reumatoide/patología , Azetidinas/uso terapéutico , Biomarcadores/metabolismo , Femenino , Expresión Génica , Humanos , Inflamación , Factor 1 Regulador del Interferón/genética , Interferones/genética , Inhibidores de las Cinasas Janus/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Piperidinas/uso terapéutico , Purinas , Pirazoles , Pirimidinas/uso terapéutico , Pirroles/uso terapéutico , Sulfonamidas/uso terapéutico , Membrana Sinovial/inmunología , Membrana Sinovial/metabolismo , Membrana Sinovial/patología , Sinoviocitos/metabolismo , Factor de Necrosis Tumoral alfa/genética
10.
PLoS One ; 14(7): e0219698, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31344123

RESUMEN

The era of next-generation sequencing has mounted the foundation of many gene expression studies. In rheumatoid arthritis research, this has led to the discovery of important candidate genes which offered novel insights into mechanisms and their possible roles in the cure of the disease. In the last years, data generation has outstripped data analysis and while many studies focused on specific aspects of the disease, a global picture of the disease is not yet accomplished. Here, we analyzed and compared a collection of gene expression information from healthy individuals and from patients suffering under different arthritis conditions from published studies containing the following clinical conditions: early and established rheumatoid arthritis, osteoarthritis and arthralgia. We show comprehensive overviews of this data collection and give new insights specifically on gene expression in the early stage, into sex-dependent gene expression, and we describe general differences in expression of different biotypes of genes. Many genes that are related to cytoskeleton changes (actin filament related genes) are differently expressed in early rheumatoid arthritis in comparison to healthy subjects; interestingly, eight of these genes reverse their expression ratio significantly between men and women compared early rheumatoid arthritis and healthy subjects. There are some slighter changes between men and woman between the conditions early and established rheumatoid arthritis. Another aspect are miRNAs and other gene biotypes which are not only promising candidates for diagnoses but also change their expression grossly in average at rheumatoid arthritis and arthralgia compared to the healthy condition. With a selection of intersecting genes, we were able to generate simple classification models to distinguish between healthy and rheumatoid arthritis as well as between early rheumatoid arthritis to other arthritides based on gene expression.


Asunto(s)
Artritis Reumatoide/genética , Regulación de la Expresión Génica , Sexismo , Análisis por Conglomerados , Femenino , Ontología de Genes , Humanos , Masculino , Modelos Biológicos , Osteoartritis/genética , Análisis de Componente Principal
11.
Lab Invest ; 99(5): 648-658, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30679758

RESUMEN

Fibroblast-like synoviocytes (FLS) are major contributors to joint inflammation in rheumatoid arthritis (RA). Forkhead box O 3 (FOXO3) perturbations in immune cells are increasingly linked to RA pathogenesis. Here, we show that FOXO3 is distinctly inactivated/phosphorylated in the FLS of rheumatoid synovitis. In vitro, stimulation of FLS with tumor necrosis factor-alpha α (TNFα) induced a rapid and sustained inactivation of FOXO3. mRNA profiling revealed that the inactivation of FOXO3 is important for the sustained pro-inflammatory interferon response to TNFα (CXCL9, CXCL10, CXCL11, and TNFSF18). Mechanistically, our studies demonstrate that the inactivation of FOXO3 results from TNF-induced downregulation of phosphoinositide-3-kinase-interacting protein 1 (PIK3IP1). Thus, we identified FOXO3 and its modulator PIK3IP1 as a critical regulatory circuit for the inflammatory response of the resident mesenchymal cells to TNFα and contribute insight into how the synovial tissue brings about chronic inflammation that is driven by TNFα.


Asunto(s)
Fibroblastos/efectos de los fármacos , Proteína Forkhead Box O3/genética , Inflamación/genética , Sinoviocitos/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Adulto , Anciano , Anciano de 80 o más Años , Artritis Reumatoide/genética , Artritis Reumatoide/metabolismo , Artritis Reumatoide/patología , Células Cultivadas , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Proteína Forkhead Box O3/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Persona de Mediana Edad , Sinoviocitos/citología , Sinoviocitos/metabolismo
12.
Ann Rheum Dis ; 77(10): 1490-1497, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29959183

RESUMEN

OBJECTIVES: Bone destruction in rheumatoid arthritis is mediated by osteoclasts (OC), which are derived from precursor cells of the myeloid lineage. The role of the two monocyte subsets, classical monocytes (expressing CD115, Ly6C and CCR2) and non-classical monocytes (which are CD115 positive, but low in Ly6C and CCR2), in serving as precursors for OC in arthritis is still elusive. METHODS: We investigated CCR2-/- mice, which lack circulating classical monocytes, crossed into hTNFtg mice for the extent of joint damage. We analysed monocyte subsets in hTNFtg and K/BxN serum transfer arthritis by flow cytometry. We sorted monocyte subsets and analysed their potential to differentiate into OC and their transcriptional response in response to RANKL by RNA sequencing. With these data, we performed a gene ontology enrichment analysis and gene set enrichment analysis. RESULTS: We show that in hTNFtg arthritis local bone erosion and OC generation are even enhanced in the absence of CCR2. We further show the numbers of non-classical monocytes in blood are elevated and are significantly correlated with histological signs of joint destruction. Sorted non-classical monocytes display an increased capacity to differentiate into OCs. This is associated with an increased expression of signal transduction components of RANK, most importantly TRAF6, leading to an increased responsiveness to RANKL. CONCLUSION: Therefore, non-classical monocytes are pivotal cells in arthritis tissue damage and a possible target for therapeutically intervention for the prevention of inflammatory joint damage.


Asunto(s)
Artritis Experimental/fisiopatología , Artritis Reumatoide/fisiopatología , Resorción Ósea/fisiopatología , Monocitos/fisiología , Osteoclastos/fisiología , Animales , Artritis Experimental/complicaciones , Artritis Reumatoide/complicaciones , Resorción Ósea/etiología , Diferenciación Celular , Modelos Animales de Enfermedad , Citometría de Flujo , Ratones , Ligando RANK/metabolismo , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Receptores CCR2/metabolismo , Transducción de Señal/fisiología , Factor 6 Asociado a Receptor de TNF/metabolismo
13.
Cell Rep ; 23(7): 2157-2167, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29768212

RESUMEN

Accumulating evidence suggests that metabolic master regulators, including mTOR, regulate adaptive and innate immune responses. Resident mesenchymal tissue components are increasingly recognized as key effector cells in inflammation. Whether mTOR also controls the inflammatory response in fibroblasts is insufficiently studied. Here, we show that TNF signaling co-opts the mTOR pathway to shift synovial fibroblast (FLS) inflammation toward an IFN response. mTOR pathway activation is associated with decreased NF-κB-mediated gene expression (e.g., PTGS2, IL-6, and IL-8) but increased STAT1-dependent gene expression (e.g., CXCL11 and TNFSF13B). We further demonstrate how metabolic inputs, such as amino acids, impinge on TNF-mTORC1 signaling to differentially regulate pro-inflammatory signaling circuits. Our results define a critical role for mTOR in the regulation of the pro-inflammatory response in FLSs and unfold its pathogenic involvement in TNF-driven diseases, such as rheumatoid arthritis (RA).


Asunto(s)
Microambiente Celular , Fibroblastos/patología , Inflamación/patología , Sinoviocitos/metabolismo , Sinoviocitos/patología , Serina-Treonina Quinasas TOR/metabolismo , Aminoácidos/metabolismo , Artritis Reumatoide/patología , Regulación de la Expresión Génica , Humanos , Inhibidor NF-kappaB alfa/metabolismo , FN-kappa B/metabolismo , Reproducibilidad de los Resultados , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
14.
Rheumatology (Oxford) ; 57(3): 572-577, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29228301

RESUMEN

Objectives: The aim was to explore the function of the T-cell cytokine IFNγ for mesenchymal tissue remodelling in RA and to determine whether IFNγ signalling controls the invasive potential of fibroblast-like synoviocytes (FLS). Methods: To assess architectural responses, FLS were cultured in three-dimensional micromasses. FLS motility was analysed in migration and invasion assays. Signalling events relevant to cellular motility were defined by western blots. Baricitinib and small interfering RNA pools were used to suppress Janus kinase (JAK) functions. Results: Histological analyses of micromasses revealed unique effects of IFNγ on FLS shape and tissue organization. This was consistent with accelerated migration upon IFNγ stimulation. Given that cell shape and cell motility are under the control of the focal adhesion kinase (FAK), we next analysed its activity. Indeed, IFNγ stimulation induced the phosphorylation of FAK-Y925, a phosphosite implicated in FAK-mediated cell migration. Small interfering RNA knockdown of JAK2, but not JAK1, substantially abrogated FAK activation by IFNγ. Correspondingly, IFNγ-induced FAK activation and invasion of FLS was abrogated by the JAK inhibitor, baricitinib. Conclusion: Our study contributes insight into the synovial response to IFNγ and reveals JAK2 as a potential therapeutic target for FLS-mediated joint destruction in arthritis, especially in RA.


Asunto(s)
Artritis Reumatoide/metabolismo , Fibroblastos/metabolismo , Interferón gamma/fisiología , Janus Quinasa 2/antagonistas & inhibidores , Sinoviocitos/metabolismo , Adulto , Artritis Reumatoide/tratamiento farmacológico , Azetidinas/farmacología , Técnicas de Cultivo de Célula , Movimiento Celular/fisiología , Células Cultivadas , Femenino , Quinasa 1 de Adhesión Focal/fisiología , Humanos , Inhibidores de las Cinasas Janus/farmacología , Masculino , Persona de Mediana Edad , Purinas , Pirazoles , ARN Interferente Pequeño/farmacología , Sulfonamidas/farmacología
15.
Arthritis Res Ther ; 18: 63, 2016 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-26970742

RESUMEN

BACKGROUND: Our aim was to investigate the role of nicotinic acetylcholine receptors (nAChRs) in in-vitro osteoclastogenesis and in in-vivo bone homeostasis. METHODS: The presence of nAChR subunits as well as the in-vitro effects of nAChR agonists were investigated by ex vivo osteoclastogenesis assays, real-time polymerase chain reaction, Western blot and flow cytometry in murine bone marrow-derived macrophages differentiated in the presence of recombinant receptor activator of nuclear factor kappa B ligand (RANKL) and macrophage colony-stimulating factor (M-CSF). The bone phenotype of mice lacking various nAChR subunits was investigated by peripheral quantitative computed tomography and histomorphometric analysis. Oscillations in the intracellular calcium concentration were detected by measuring the Fura-2 fluorescence intensity. RESULTS: We could demonstrate the presence of several nAChR subunits in bone marrow-derived macrophages stimulated with RANKL and M-CSF, and showed that they are capable of producing acetylcholine. nAChR ligands reduced the number of osteoclasts as well as the number of tartrate-resistant acidic phosphatase-positive mononuclear cells in a dose-dependent manner. In vitro RANKL-mediated osteoclastogenesis was reduced in mice lacking α7 homomeric nAChR or ß2-containing heteromeric nAChRs, while bone histomorphometry revealed increased bone volume as well as impaired osteoclastogenesis in male mice lacking the α7 nAChR. nAChR ligands inhibited RANKL-induced calcium oscillation, a well-established phenomenon of osteoclastogenesis. This inhibitory effect on Ca(2+) oscillation subsequently led to the inhibition of RANKL-induced NFATc1 and c-fos expression after long-term treatment with nicotine. CONCLUSIONS: We have shown that the activity of nAChRs conveys a marked effect on osteoclastogenesis in mice. Agonists of these receptors inhibited calcium oscillations in osteoclasts and blocked the RANKL-induced activation of c-fos and NFATc1. RANKL-mediated in-vitro osteoclastogenesis was reduced in α7 knockout mice, which was paralleled by increased tibial bone volume in male mice in vivo.


Asunto(s)
Remodelación Ósea/fisiología , Huesos/fisiología , Osteoclastos/metabolismo , Receptores Nicotínicos/metabolismo , Animales , Western Blotting , Citometría de Flujo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa
16.
Rheumatology (Oxford) ; 55(4): 710-20, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26672908

RESUMEN

OBJECTIVE: Abatacept (CTLA-4Ig) blocks CD28-mediated T cell activation by binding to the costimulatory B7 ligands CD80/CD86 on antigen presenting cells. Costimulatory molecules, however, can also be expressed on T cells upon activation. Therefore, the aim of our study was to investigate direct effects of CTLA-4Ig on distinct T cell subsets in RA patients. METHODS: Phenotypic and functional analyses of CD4(+) T cells, including CD4(+) FoxP3(+) CD25(+) regulatory T cells (Treg), from RA patients were performed before and during CTLA-4Ig therapy. In addition T cells from healthy volunteers were analysed on in vitro culture with CTLA-4Ig or anti-CD80 and anti-CD86 antibodies. Apoptotic DNA fragmentation in CD4(+) and CD4(+) FoxP3(+) T cells was measured by TUNEL staining. RESULTS: We observed an increase in T cells, including Treg cells, after initiation of CTLA-4Ig therapy, which was linked to a downregulation of activation-associated marker molecules and CD95 on CD4(+) T cells and Treg cells. CTLA-4Ig decreased CD95-mediated cell death in vitro in a dose-dependent manner. Functional analysis of isolated Treg cells from RA patients further revealed a diminished suppression of responder T cell proliferation. This was found to be due to CTLA-4Ig-mediated blocking of CD80 and CD86 on responder T cells that led to a diminished susceptibility for Treg cell suppression. CONCLUSION: CTLA-4Ig therapy in RA patients exerts effects beyond the suppression of T cell activation, which has to be taken into account as an additional mechanism of CTLA-4Ig treatment.


Asunto(s)
Abatacept/farmacología , Antirreumáticos/farmacología , Apoptosis/efectos de los fármacos , Artritis Reumatoide/inmunología , Subgrupos de Linfocitos T/efectos de los fármacos , Linfocitos T Reguladores/efectos de los fármacos , Abatacept/uso terapéutico , Antirreumáticos/uso terapéutico , Apoptosis/inmunología , Artritis Reumatoide/tratamiento farmacológico , Linfocitos T CD4-Positivos/efectos de los fármacos , Células Cultivadas , Femenino , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Inmunofenotipificación , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Masculino , Persona de Mediana Edad , Receptor fas/inmunología
17.
Cell Rep ; 11(12): 1919-28, 2015 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-26095358

RESUMEN

Lipid metabolism and receptor-mediated signaling are highly intertwined processes that cooperate to fulfill cellular functions and safeguard cellular homeostasis. Activation of Toll-like receptors (TLRs) leads to a complex cellular response, orchestrating a diverse range of inflammatory events that need to be tightly controlled. Here, we identified the GPI-anchored Sphingomyelin Phosphodiesterase, Acid-Like 3B (SMPDL3B) in a mass spectrometry screening campaign for membrane proteins co-purifying with TLRs. Deficiency of Smpdl3b in macrophages enhanced responsiveness to TLR stimulation and profoundly changed the cellular lipid composition and membrane fluidity. Increased cellular responses could be reverted by re-introducing affected ceramides, functionally linking membrane lipid composition and innate immune signaling. Finally, Smpdl3b-deficient mice displayed an intensified inflammatory response in TLR-dependent peritonitis models, establishing its negative regulatory role in vivo. Taken together, our results identify the membrane-modulating enzyme SMPDL3B as a negative regulator of TLR signaling that functions at the interface of membrane biology and innate immunity.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/genética , Inmunidad Innata/genética , Inflamación/genética , Peritonitis/genética , Animales , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/inmunología , Modelos Animales de Enfermedad , Humanos , Inflamación/inmunología , Inflamación/patología , Lípidos/inmunología , Macrófagos/inmunología , Ratones , Peritonitis/inmunología , Peritonitis/patología , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología
18.
Eur J Immunol ; 45(5): 1500-11, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25678110

RESUMEN

Secretion of type I interferon (IFN) is the first cellular reaction to invading pathogens. Despite the protective function of these cytokines, an excessive response to their action can contribute to serious pathologies, such as autoimmune diseases. Transcripts of most cytokines contain adenylate-uridylate (A/U)-rich elements (AREs) that make them highly unstable. RNA-binding proteins (RBPs) are mediators of the regulatory mechanisms that determine the fate of mRNAs containing AREs. Here, we applied an affinity proteomic approach and identified lethal, abnormal vision, drosophila-like 1 (ELAVL1)/Hu antigen R (HuR) as the predominant RBP of the IFN-ß mRNA ARE. Reduced expression or chemical inhibition of HuR severely hampered the type I IFN response in various cell lines and fibroblast-like synoviocytes isolated from joints of rheumatoid arthritis patients. These results define a role for HuR as a potent modulator of the type I IFN response. Taken together, HuR could be used as therapeutic target for diseases where type I IFN production is exaggerated.


Asunto(s)
Proteínas ELAV/inmunología , Interferón Tipo I/biosíntesis , Interferón beta/genética , Elementos Ricos en Adenilato y Uridilato , Artritis Reumatoide/genética , Artritis Reumatoide/inmunología , Secuencia de Bases , Proteínas ELAV/antagonistas & inhibidores , Proteínas ELAV/genética , Proteína 1 Similar a ELAV , Células HeLa , Humanos , Inductores de Interferón/farmacología , Datos de Secuencia Molecular , Poli I-C/farmacología , Multimerización de Proteína , Procesamiento Postranscripcional del ARN/efectos de los fármacos , Estabilidad del ARN/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Membrana Sinovial/inmunología
19.
Wien Med Wochenschr ; 165(1-2): 40-5, 2015 Jan.
Artículo en Alemán | MEDLINE | ID: mdl-25411010

RESUMEN

Biologics have become indispensable in the last decade in the treatment of the more common rheumatic diseases. For treating systemic lupus erythematodes (SLE), B-cell depletion, albeit off-label, has been a well-accepted strategy in severe and refractory disease. Unexpectedly, however, the results of the first randomized controlled rituximab trials in SLE were negative. New trials with improved study protocols are ongoing, which should resolve this issue. In 2012, with the approval of belimumab, SLE finally entered the era of approved biological therapies. The anti-Blys/BAFF antibody belimumab showed prevention of SLE flares, glucocorticoid sparing, and significant improvement in the quality of life of SLE patients, in part by drastically reducing immune complex mediated fatigue. Positive reports on further targeting approaches give hope that additional biological agents will be available for SLE therapy soon.


Asunto(s)
Antirreumáticos/uso terapéutico , Productos Biológicos/uso terapéutico , Lupus Eritematoso Sistémico/tratamiento farmacológico , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/uso terapéutico , Linfocitos B/efectos de los fármacos , Productos Biológicos/efectos adversos , Aprobación de Drogas , Humanos , Lupus Eritematoso Sistémico/inmunología , Uso Fuera de lo Indicado , Ensayos Clínicos Controlados Aleatorios como Asunto , Rituximab/efectos adversos , Rituximab/uso terapéutico
20.
Arthritis Res Ther ; 16(2): R104, 2014 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-24774748

RESUMEN

INTRODUCTION: Systemic lupus erythematosus (SLE) is a heterogenous autoimmune disease, which can affect different organs. Increased proportions of CD4⁺CD25-Foxp3⁺ T cells have been described in SLE patients. The exact role of this cell population in SLE patients still remains unclear. We therefore analyzed this T cell subset in a large cohort of SLE patients with different organ manifestations. METHODS: Phenotypic analyses, proportions and absolute cell numbers of CD4⁺CD25-Foxp3⁺ T cells were determined by flow cytometry (FACS) in healthy controls (HC) (n = 36) and SLE patients (n = 61) with different organ manifestations. CD4⁺CD25⁻Foxp3⁺ T cells were correlated with clinical data, the immunosuppressive therapy and different disease activity indices. In patients with active glomerulonephritis, CD4⁺CD25⁻Foxp3⁺ T cells were analyzed in urine sediment samples. Time course analyses of CD4⁺CD25⁻Foxp3⁺ T cells were performed in patients with active disease activity before and after treatment with cyclophosphamide and prednisone. RESULTS: CD4⁺CD25⁻Foxp3⁺ T cells were significantly increased in active SLE patients and the majority expressed Helios. Detailed analysis of this patient cohort revealed increased proportions of CD4⁺CD25⁻Foxp3⁺ T cells in SLE patients with renal involvement. CD4⁺CD25⁻Foxp3⁺ T cells were also detected in urine sediment samples of patients with active glomerulonephritis and correlated with the extent of proteinuria. CONCLUSION: CD4⁺CD25⁻Foxp3⁺ T cells resemble regulatory rather than activated T cells. Comparative analysis of CD4⁺CD25⁻Foxp3⁺ T cells in SLE patients revealed a significant association of this newly described cell population with active nephritis. Therefore CD4⁺CD25⁻Foxp3⁺ T cells might serve as an important tool to recognize and monitor SLE patients with renal involvement.


Asunto(s)
Biomarcadores/sangre , Linfocitos T CD4-Positivos/inmunología , Nefritis Lúpica/inmunología , Subgrupos de Linfocitos T/inmunología , Adulto , Femenino , Citometría de Flujo , Factores de Transcripción Forkhead/inmunología , Humanos , Inmunofenotipificación , Masculino , Persona de Mediana Edad , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...