Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 134(10)2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38747285

RESUMO

Transforming growth factor ß (TGF-ß) signaling is a core pathway of fibrosis, but the molecular regulation of the activation of latent TGF-ß remains incompletely understood. Here, we demonstrate a crucial role of WNT5A/JNK/ROCK signaling that rapidly coordinates the activation of latent TGF-ß in fibrotic diseases. WNT5A was identified as a predominant noncanonical WNT ligand in fibrotic diseases such as systemic sclerosis, sclerodermatous chronic graft-versus-host disease, and idiopathic pulmonary fibrosis, stimulating fibroblast-to-myofibroblast transition and tissue fibrosis by activation of latent TGF-ß. The activation of latent TGF-ß requires rapid JNK- and ROCK-dependent cytoskeletal rearrangements and integrin αV (ITGAV). Conditional ablation of WNT5A or its downstream targets prevented activation of latent TGF-ß, rebalanced TGF-ß signaling, and ameliorated experimental fibrosis. We thus uncovered what we believe to be a novel mechanism for the aberrant activation of latent TGF-ß in fibrotic diseases and provided evidence for targeting WNT5A/JNK/ROCK signaling in fibrotic diseases as a new therapeutic approach.


Assuntos
Fibroblastos , Fibrose , Fator de Crescimento Transformador beta , Proteína Wnt-5a , Quinases Associadas a rho , Proteína Wnt-5a/metabolismo , Proteína Wnt-5a/genética , Animais , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/genética , Camundongos , Humanos , Fibroblastos/metabolismo , Fibroblastos/patologia , Quinases Associadas a rho/metabolismo , Quinases Associadas a rho/genética , Escleroderma Sistêmico/patologia , Escleroderma Sistêmico/metabolismo , Escleroderma Sistêmico/genética , Camundongos Knockout , Proteínas Wnt/metabolismo , Proteínas Wnt/genética , Sistema de Sinalização das MAP Quinases , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Transdução de Sinais , Fibrose Pulmonar Idiopática/patologia , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/genética
2.
Ann Rheum Dis ; 2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38594058

RESUMO

BACKGROUND: The interleukin (IL)-1 receptor accessory protein (IL1RAP) is an essential coreceptor required for signalling through the IL-1, IL-33 and IL-36 receptors. Here, we investigate the antifibrotic potential of the combined inhibition of these cytokines by an anti-IL1RAP antibody to provide a scientific background for clinical development in systemic sclerosis (SSc). METHODS: The expression of IL1RAP-associated signalling molecules was determined by data mining of publicly available RNA sequencing (RNAseq) data as well as by imaging mass cytometry. The efficacy of therapeutic dosing of anti-IL1RAP antibodies was determined in three complementary mouse models: sclerodermatous chronic graft-versus-host disease (cGvHD), bleomycin-induced dermal fibrosis model and topoisomerase-I (topo)-induced fibrosis. RESULTS: SSc skin showed upregulation of IL1RAP and IL1RAP-related signalling molecules on mRNA and protein level compared with normal skin. IL-1, IL-33 and IL-36 all regulate distinct gene sets related to different pathophysiological processes in SSc. The responses of human fibroblasts and endothelial cells to IL-1, IL-33 and IL-36 were completely blocked by treatment with an anti-IL1RAP antibody in vitro. Moreover, anti-IL1RAP antibody treatment reduced dermal and pulmonary fibrosis in cGvHD-induced, bleomycin-induced and topoisomerase-induced fibrosis. Importantly, RNAseq analyses revealed effects of IL1RAP inhibition on multiple processes related to inflammation and fibrosis that are also deregulated in human SSc skin. CONCLUSION: This study provides the first evidence for the therapeutic benefits of targeting IL1RAP in SSc. Our findings have high translational potential as the anti-IL1RAP antibody CAN10 has recently entered a phase one clinical trial.

4.
J Clin Invest ; 130(9): 4888-4905, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32603313

RESUMO

TGF-ß is a master regulator of fibrosis, driving the differentiation of fibroblasts into apoptosis-resistant myofibroblasts and sustaining the production of extracellular matrix (ECM) components. Here, we identified the nuclear long noncoding RNA (lncRNA) H19X as a master regulator of TGF-ß-driven tissue fibrosis. H19X was consistently upregulated in a wide variety of human fibrotic tissues and diseases and was strongly induced by TGF-ß, particularly in fibroblasts and fibroblast-related cells. Functional experiments following H19X silencing revealed that H19X was an obligatory factor for TGF-ß-induced ECM synthesis as well as differentiation and survival of ECM-producing myofibroblasts. We showed that H19X regulates DDIT4L gene expression, specifically interacting with a region upstream of the DDIT4L gene and changing the chromatin accessibility of a DDIT4L enhancer. These events resulted in transcriptional repression of DDIT4L and, in turn, in increased collagen expression and fibrosis. Our results shed light on key effectors of TGF-ß-induced ECM remodeling and fibrosis.


Assuntos
Matriz Extracelular/metabolismo , Miofibroblastos/metabolismo , Fibrose Pulmonar/metabolismo , RNA Longo não Codificante/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Linhagem Celular , Matriz Extracelular/genética , Matriz Extracelular/patologia , Humanos , Camundongos , Miofibroblastos/patologia , Fibrose Pulmonar/genética , Fibrose Pulmonar/patologia , RNA Longo não Codificante/genética , Fator de Crescimento Transformador beta/genética
5.
J Clin Invest ; 130(5): 2347-2363, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31990678

RESUMO

Fibroblasts are key effector cells in tissue remodeling. They remain persistently activated in fibrotic diseases, resulting in progressive deposition of extracellular matrix. Although fibroblast activation may be initiated by external factors, prolonged activation can induce an "autonomous," self-maintaining profibrotic phenotype in fibroblasts. Accumulating evidence suggests that epigenetic alterations play a central role in establishing this persistently activated pathologic phenotype of fibroblasts. We demonstrated that in fibrotic skin of patients with systemic sclerosis (SSc), a prototypical idiopathic fibrotic disease, TGF-ß induced the expression of DNA methyltransferase 3A (DNMT3A) and DNMT1 in fibroblasts in a SMAD-dependent manner to silence the expression of suppressor of cytokine signaling 3 (SOCS3) by promoter hypermethylation. Downregulation of SOCS3 facilitated activation of STAT3 to promote fibroblast-to-myofibroblast transition, collagen release, and fibrosis in vitro and in vivo. Reestablishment of the epigenetic control of STAT3 signaling by genetic or pharmacological inactivation of DNMT3A reversed the activated phenotype of SSc fibroblasts in tissue culture, inhibited TGF-ß-dependent fibroblast activation, and ameliorated experimental fibrosis in murine models. These findings identify a pathway of epigenetic imprinting of fibroblasts in fibrotic disease with translational implications for the development of targeted therapies in fibrotic diseases.


Assuntos
Epigênese Genética , Miofibroblastos/metabolismo , Fator de Transcrição STAT3/metabolismo , Escleroderma Sistêmico/metabolismo , Transdução de Sinais , Proteína 3 Supressora da Sinalização de Citocinas/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , DNA (Citosina-5-)-Metiltransferase 1/biossíntese , DNA (Citosina-5-)-Metiltransferases/biossíntese , DNA Metiltransferase 3A , Feminino , Fibrose , Regulação Enzimológica da Expressão Gênica , Humanos , Masculino , Camundongos , Miofibroblastos/patologia , Escleroderma Sistêmico/patologia
6.
J Scleroderma Relat Disord ; 5(2 Suppl): 17-30, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35382225

RESUMO

Systemic sclerosis is a fibrosing chronic connective tissue disease of unknown etiology. A major hallmark of systemic sclerosis is the uncontrolled and persistent activation of fibroblasts, which release excessive amounts of extracellular matrix, lead to organ dysfunction, and cause high mobility and motility of patients. Systemic sclerosis-associated interstitial lung disease is one of the most common fibrotic organ manifestations in systemic sclerosis and a major cause of death. Treatment options for systemic sclerosis-associated interstitial lung disease and other fibrotic manifestations, however, remain very limited. Thus, there is a huge medical need for effective therapies that target tissue fibrosis, vascular alterations, inflammation, and autoimmune disease in systemic sclerosis-associated interstitial lung disease. In this review, we discuss data suggesting therapeutic ways to target different genes in distinct tissues/organs that contribute to the development of SSc.

7.
J Scleroderma Relat Disord ; 4(3): 212-218, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35382502

RESUMO

Interstitial lung disease is a common manifestation of systemic sclerosis. Systemic sclerosis-associated interstitial lung disease is characterized by progressive pulmonary fibrosis and a reduction in pulmonary function. Effective treatments for systemic sclerosis-associated interstitial lung disease are lacking. In addition to clinical similarities, systemic sclerosis-associated interstitial lung disease shows similarities to idiopathic pulmonary fibrosis in the pathophysiology of the underlying fibrotic processes. Idiopathic pulmonary fibrosis and systemic sclerosis-associated interstitial lung disease culminate in a self-sustaining pathway of pulmonary fibrosis in which fibroblasts are activated, myofibroblasts accumulate, and the excessive extracellular matrix is deposited. Nintedanib is a tyrosine kinase inhibitor that has been approved for the treatment of idiopathic pulmonary fibrosis. In patients with idiopathic pulmonary fibrosis, nintedanib slows disease progression by decreasing the rate of lung function decline. In this review, we summarize the antifibrotic, anti-inflammatory, and attenuated vascular remodeling effects of nintedanib demonstrated in in vitro studies and in animal models of aspects of systemic sclerosis. Nintedanib interferes at multiple critical steps in the pathobiology of systemic sclerosis-associated interstitial lung disease, providing a convincing rationale for its investigation as a potential therapy. Finally, we summarize the design of the randomized placebo-controlled SENSCIS® trial that is evaluating the efficacy and safety of nintedanib in patients with systemic sclerosis-associated interstitial lung disease.

8.
Epigenomics ; 9(4): 463-477, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28343418

RESUMO

Prolonged activation of fibroblasts is a central hallmark of fibrosing disorders such as systemic sclerosis (SSc). Fibroblasts are the key effector cells. They differentiate into an activated myofibroblast phenotype. In contrast to normal wound healing with transient activation, myofibroblasts persist in fibrosing disorders. Current hypothesis suggests that profibrotic cytokines might trigger epigenetic changes which contribute to the persistently activated fibroblast phenotype. In the last years, several epigenetic alterations have been described in SSc and have been linked to different pathogenic aspects of the disease, in particular to aberrant fibroblast activation and tissue fibrosis, but also to vascular manifestations and inflammation. The focus of this review is the current knowledge on epigenetic changes in fibroblast activation in SSc.


Assuntos
Epigênese Genética , Redes Reguladoras de Genes , Escleroderma Sistêmico/patologia , Animais , Citocinas/metabolismo , Metilação de DNA , Fibrose , Código das Histonas , Humanos , Miofibroblastos/imunologia , Miofibroblastos/patologia , Escleroderma Sistêmico/genética , Escleroderma Sistêmico/imunologia
9.
Ann Rheum Dis ; 75(11): 2037-2044, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26851274

RESUMO

OBJECTIVES: Notch ligands and receptors have recently been shown to be differentially expressed in osteoarthritis (OA). We aim to further elucidate the functional role of Notch signalling in OA using Notch1 antisense transgenic (Notch1 AS) mice. METHODS: Notch and hedgehog signalling were analysed by real-time PCR and immunohistochemistry. Notch-1 AS mice were employed as a model of impaired Notch signalling in vivo. Experimental OA was induced by destabilisation of the medial meniscus (DMM). The extent of cartilage destruction and osteophyte formation was analysed by safranin-O staining with subsequent assessment of the Osteoarthritis Research Society International (OARSI) and Mankin scores and µCT scanning. Collagen X staining was used as a marker of chondrocyte hypertrophy. The role of hairy/enhancer of split 1 (Hes-1) was investigated with knockdown and overexpression experiments. RESULTS: Notch signalling was activated in human and murine OA with increased expression of Jagged1, Notch-1, accumulation of the Notch intracellular domain 1 and increased transcription of Hes-1. Notch1 AS mice showed exacerbated OA with increases in OARSI scores, osteophyte formation, increased subchondral bone plate density, collagen X and osteocalcin expression and elevated levels of Epas1 and ADAM-TS5 mRNA. Inhibition of the Notch pathway induced activation of hedgehog signalling with induction of Gli-1 and Gli-2 and increased transcription of hedgehog target genes. The regulatory effects of Notch signalling on Gli-expression were mimicked by Hes-1. CONCLUSIONS: Inhibition of Notch signalling activates hedgehog signalling, enhances chondrocyte hypertrophy and exacerbates experimental OA including osteophyte formation. These data suggest that the activation of the Notch pathway may limit aberrant hedgehog signalling in OA.


Assuntos
Artrite Experimental/metabolismo , Proteínas de Transporte/metabolismo , Condrócitos/metabolismo , Glicoproteínas de Membrana/metabolismo , Osteoartrite/metabolismo , Receptor Notch1/metabolismo , Fatores de Transcrição HES-1/metabolismo , Animais , Cartilagem Articular/metabolismo , Camundongos , Camundongos Transgênicos , Osteófito/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Índice de Gravidade de Doença , Transdução de Sinais
10.
Ann Rheum Dis ; 75(6): 1203-10, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26113650

RESUMO

OBJECTIVES: Autophagy has recently been shown to regulate osteoclast activity and osteoclast differentiation. Here, we aim to investigate the impact of autophagy inhibition as a potential therapeutic approach for the treatment of osteoporosis in preclinical models. METHODS: Systemic bone loss was induced in mice by glucocorticoids and by ovariectomy (OVX). Autophagy was targeted by conditional inactivation of autophagy-related gene 7 (Atg7) and by treatment with chloroquine (CQ). Bone density was evaluated by microCT. The role of autophagy on osteoclastogenesis was analysed by osteoclastogenesis and bone resorption assays. The quantification of receptor activator of nuclear factor κ B ligand and osteoprotegerin proteins in cocultures was performed using ELISA whereas that of osteoclast and osteoblast differentiation markers was by qPCR. RESULTS: Selective deletion of Atg7 in monocytes from Atg7(fl/fl)_x_LysM-Cre mice mitigated glucocorticoid-induced and OVX-induced osteoclast differentiation and bone loss compared with Atg7(fl/fl) littermates. Pharmacological inhibition of autophagy by treatment with CQ suppressed glucocorticoid-induced osteoclastogenesis and protected mice from bone loss. Similarly, inactivation of autophagy shielded mice from OVX-induced bone loss. Inhibition of autophagy led to decreased osteoclast differentiation with lower expression of osteoclast markers such as NFATc1, tartrate-resistant acid phosphatase, OSCAR and cathepsin K and attenuated bone resorption in vitro. In contrast, osteoblast differentiation was not affected by inhibition of autophagy. CONCLUSIONS: Pharmacological or genetic inactivation of autophagy ameliorated glucocorticoid-induced and OVX-induced bone loss by inhibiting osteoclastogenesis. These findings may have direct translational implications for the treatment of osteoporosis, since inhibitors of autophagy such as CQ are already in clinical use.


Assuntos
Autofagia/efeitos dos fármacos , Osteoporose/prevenção & controle , Animais , Proteína 7 Relacionada à Autofagia/genética , Células Cultivadas , Cloroquina/farmacologia , Cloroquina/uso terapêutico , Modelos Animais de Doenças , Feminino , Deleção de Genes , Glucocorticoides , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Terapia de Alvo Molecular , Monócitos/metabolismo , Osteoclastos/efeitos dos fármacos , Osteoclastos/fisiologia , Osteogênese/efeitos dos fármacos , Osteoporose/induzido quimicamente , Osteoporose/etiologia , Osteoporose/patologia , Ovariectomia
11.
Ann Rheum Dis ; 75(8): 1521-6, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26338038

RESUMO

OBJECTIVES: TYK2 is a common genetic risk factor for several autoimmune diseases. This gene encodes a protein kinase involved in interleukin 12 (IL-12) pathway, which is a well-known player in the pathogenesis of systemic sclerosis (SSc). Therefore, we aimed to assess the possible role of this locus in SSc. METHODS: This study comprised a total of 7103 patients with SSc and 12 220 healthy controls of European ancestry from Spain, USA, Germany, the Netherlands, Italy and the UK. Four TYK2 single-nucleotide polymorphisms (V362F (rs2304256), P1104A (rs34536443), I684S (rs12720356) and A928V (rs35018800)) were selected for follow-up based on the results of an Immunochip screening phase of the locus. Association and dependence analyses were performed by the means of logistic regression and conditional logistic regression. Meta-analyses were performed using the inverse variance method. RESULTS: Genome-wide significance level was reached for TYK2 V362F common variant in our pooled analysis (p=3.08×10(-13), OR=0.83), while the association of P1104A, A928V and I684S rare and low-frequency missense variants remained significant with nominal signals (p=2.28×10(-3), OR=0.80; p=1.27×10(-3), OR=0.59; p=2.63×10(-5), OR=0.83, respectively). Interestingly, dependence and allelic combination analyses showed that the strong association observed for V362F with SSc, corresponded to a synthetic association dependent on the effect of the three previously mentioned TYK2 missense variants. CONCLUSIONS: We report for the first time the association of TYK2 with SSc and reinforce the relevance of the IL-12 pathway in SSc pathophysiology.


Assuntos
Interleucina-12/fisiologia , Polimorfismo de Nucleotídeo Único , Escleroderma Sistêmico/genética , TYK2 Quinase/genética , Estudos de Casos e Controles , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Humanos , Mutação de Sentido Incorreto , Escleroderma Sistêmico/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia
12.
Ann Rheum Dis ; 73(6): 1264-8, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24445254

RESUMO

OBJECTIVES: The morphogen pathways Hedgehog, Wnt and Notch are attractive targets for antifibrotic therapies in systemic sclerosis. Interference with stem cell regeneration, however, may complicate the use of morphogen pathway inhibitors. We therefore tested the hypothesis that combination therapies with low doses of Hedgehog, Wnt and Notch inhibitors maybe safe and effective for the treatment of fibrosis. METHODS: Skin fibrosis was induced by bleomycin and by overexpression of a constitutively active TGF-ß receptor type I. Adverse events were assessed by clinical monitoring, pathological evaluation and quantification of Lgr5-positive intestinal stem cells. RESULTS: Inhibition of Hedgehog, Wnt and Notch signalling dose-dependently ameliorated bleomycin-induced and active TGF-ß receptor type I-induced fibrosis. Combination therapies with low doses of Hedgehog/Wnt inhibitors or Hedgehog/Notch inhibitors demonstrated additive antifibrotic effects in preventive as well as in therapeutic regimes. Combination therapies were well tolerated. In contrast with high dose monotherapies, combination therapies did not reduce the number of Lgr5 positive intestinal stem cells. CONCLUSIONS: Combined inhibition of morphogen pathways exerts additive antifibrotic effects. Combination therapies are well tolerated and, in contrast to high dose monotherapies, may not impair stem cell renewal. Combined targeting of morphogen pathways may thus help to overcome dose-limiting toxicity of Hedgehog, Wnt and Notch signalling.


Assuntos
Fibrose/tratamento farmacológico , Proteínas Hedgehog/antagonistas & inibidores , Receptores Notch/antagonistas & inibidores , Escleroderma Sistêmico/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Pele/efeitos dos fármacos , Proteínas Wnt/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Animais , Bleomicina , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Modelos Animais de Doenças , Quimioterapia Combinada , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Proteínas Serina-Treonina Quinases/genética , Pirimidinonas/farmacologia , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/genética , Alcaloides de Veratrum/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos
13.
J Bone Miner Res ; 27(12): 2442-51, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22806960

RESUMO

Osteoporosis is characterized by enhanced differentiation of bone-resorbing osteoclasts, resulting in a rapid loss of functional trabecular bone. Bone-forming osteoblasts and osteoblast-derived osteocytes perform a key role in the regulation of osteoclast development by providing both the pro-osteoclastogenic cytokine receptor activator of NF-κB ligand (RANKL) and its natural decoy receptor osteoprotegerin (OPG). By regulating the RANKL/OPG ratio, osteoblasts hence determine the rate of both osteoclast differentiation and bone turnover. Here, we describe a novel role for liver X receptors (LXRs) during the crosstalk of bone-forming osteoblasts and bone-resorbing osteoclasts. By using a system of osteoblast/osteoclast cocultures, we identify LXRs as regulator of RANKL expression and the RANKL/OPG ratio in osteoblasts. Activation of LXRs drastically reduced the RANKL/OPG ratio and interfered with osteoblast-mediated osteoclast differentiation in vitro. During an ovariectomy (OVX)-induced model of postmenopausal osteoporosis, the application of an LXR agonist shifted the RANKL/OPG ratio in vivo, ameliorated the enhanced osteoclast differentiation, and provided complete protection from OVX-induced bone loss. These results reveal an unexpected involvement of LXRs in the regulation of bone turnover and highlight a potential role for LXRs as novel targets in the treatment of osteoporosis and related diseases.


Assuntos
Receptores Nucleares Órfãos/fisiologia , Osteoblastos/fisiologia , Osteoclastos/fisiologia , Animais , Benzoatos/farmacologia , Benzilaminas/farmacologia , Reabsorção Óssea/tratamento farmacológico , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Feminino , Humanos , Receptores X do Fígado , Camundongos , Receptores Nucleares Órfãos/agonistas , Receptores Nucleares Órfãos/uso terapêutico , Osteoblastos/citologia , Osteoporose/tratamento farmacológico , Osteoprotegerina/metabolismo , Ovariectomia , Ligante RANK/metabolismo
14.
F1000 Med Rep ; 12009 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-20948682

RESUMO

In recent years, many potential antifibrotic treatment strategies have emerged from molecular studies of systemic sclerosis. Few biologicals have already entered clinical trials and these may hopefully prove to be effective in this progressive, profibrotic disease.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA