Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cancer Immunol Immunother ; 73(2): 34, 2024 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-38280067

RESUMO

Immune checkpoint inhibitors (ICIs) have demonstrated efficacy and improved survival in a growing number of cancers. Despite their success, ICIs are associated with immune-related adverse events that can interfere with their use. Therefore, safer approaches are needed. CD6, expressed by T-lymphocytes and human NK cells, engages in cell-cell interactions by binding to its ligands CD166 (ALCAM) and CD318 (CDCP1). CD6 is a target protein for regulating immune responses and is required for the development of several mouse models of autoimmunity. Interestingly, CD6 is exclusively expressed on immune cells while CD318 is strongly expressed on most cancers. Here we demonstrate that disrupting the CD6-CD318 axis with UMCD6, an anti-CD6 monoclonal antibody, prolongs survival of mice in xenograft mouse models of human breast and prostate cancer, treated with infusions of human lymphocytes. Analysis of tumor-infiltrating immune cells showed that augmentation of lymphocyte cytotoxicity by UMCD6 is due to effects of this antibody on NK, NKT and CD8 + T cells. In particular, tumor-infiltrating cytotoxic lymphocytes from UMCD6-treated mice expressed higher levels of perforin and were found in higher proportions than those from IgG-treated mice. Moreover, RNA-seq analysis of human NK-92 cells treated with UMCD6 revealed that UMCD6 up-regulates the NKG2D-DAP10 receptor complex, important in NK cell activation, as well as its downstream target PI3K. Our results now describe the phenotypic changes that occur on immune cells upon treatment with UMCD6 and further confirm that the CD6-CD318 axis can regulate the activation state of cytotoxic lymphocytes and their positioning within the tumor microenvironment.


Assuntos
Antineoplásicos , Neoplasias , Animais , Humanos , Camundongos , Anticorpos Monoclonais/farmacologia , Antígenos CD , Antígenos de Diferenciação de Linfócitos T/metabolismo , Antígenos de Neoplasias , Moléculas de Adesão Celular , Linfócitos/metabolismo , Microambiente Tumoral
2.
Int J Mol Sci ; 25(5)2024 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-38473703

RESUMO

Multiple sclerosis (MS) is the most common autoimmune demyelinating disease of the central nervous system (CNS), consisting of heterogeneous clinical courses varying from relapsing-remitting MS (RRMS), in which disability is linked to bouts of inflammation, to progressive disease such as primary progressive MS (PPMS) and secondary progressive MS (SPMS), in which neurological disability is thought to be linked to neurodegeneration. As a result, successful therapeutics for progressive MS likely need to have both anti-inflammatory and direct neuroprotective properties. The modulation of sphingosine-1-phosphate (S1P) receptors has been implicated in neuroprotection in preclinical animal models. Siponimod/BAF312, the first oral treatment approved for SPMS, may have direct neuroprotective benefits mediated by its activity as a selective (S1P receptor 1) S1P1 and (S1P receptor 5) S1P5 modulator. We showed that S1P1 was mainly present in cortical neurons in lesioned areas of the MS brain. To gain a better understanding of the neuroprotective effects of siponimod in MS, we used both rat neurons and human-induced pluripotent stem cell (iPSC)-derived neurons treated with the neuroinflammatory cytokine tumor necrosis factor-alpha (TNF-α). Cell survival/apoptotic assays using flow cytometry and IncuCyte live cell analyses showed that siponimod decreased TNF-α induced neuronal cell apoptosis in both rat and human iPSCs. Importantly, a transcriptomic analysis revealed that mitochondrial oxidative phosphorylation, NFκB and cytokine signaling pathways contributed to siponimod's neuroprotective effects. Our data suggest that the neuroprotection of siponimod/BAF312 likely involves the relief of oxidative stress in neuronal cells. Further studies are needed to explore the molecular mechanisms of such interactions to determine the relationship between mitochondrial dysfunction and neuroinflammation/neurodegeneration.


Assuntos
Azetidinas , Compostos de Benzil , Esclerose Múltipla Crônica Progressiva , Esclerose Múltipla , Fármacos Neuroprotetores , Humanos , Animais , Ratos , Receptores de Esfingosina-1-Fosfato , Doenças Neuroinflamatórias , Fármacos Neuroprotetores/farmacologia , Fator de Necrose Tumoral alfa/uso terapêutico , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla Crônica Progressiva/tratamento farmacológico , Morte Celular
3.
Curr Opin Rheumatol ; 34(6): 343-350, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-35979691

RESUMO

PURPOSE OF REVIEW: Systemic sclerosis (SSc) is a chronic rheumatic disease that is characterized by immune activation, vasculopathy and fibrosis of the skin and internal organs. It has been proposed that premature onset of ageing pathways and associated senescent changes in cells contribute to the clinical and pathological features of SSc. The aim of this review is to critically review recent insights into the involvement of cellular senescence in SSc. RECENT FINDINGS: Cellular senescence plays a critical role in SSc pathogenesis, particularly involving endothelial cells and fibroblasts. Immunosenescence could also contribute to SSc pathogenesis by direct alteration of cellular functions or indirect promotion of defective immune surveillance. Molecular studies have shed some light on how cellular senescence contributes to fibrosis. Recent and planned proof-of-concept trials using senotherapeutics showed promising results in fibrotic diseases, including SSc. SUMMARY: There is increasing evidence implicating cellular senescence in SSc. The mechanisms underlying premature cellular senescence in SSc, and its potential role in pathogenesis, merit further investigation. Emerging drugs targeting senescence-related pathways might be potential therapeutic options for SSc.


Assuntos
Células Endoteliais , Escleroderma Sistêmico , Senescência Celular , Fibroblastos , Fibrose , Humanos
4.
Proc Natl Acad Sci U S A ; 116(9): 3695-3702, 2019 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-30755532

RESUMO

Scleroderma (SSc) is a complex disease that involves activation of the immune system, vascular complications, and tissue fibrosis. The histone methyltransferase enhancer of zeste homolog 2 (EZH2) mediates trimethylation of lysine 27 of histone 3 (H3K27me3), which acts as a repressive epigenetic mark. Both EZH2 and H3K27me3 were elevated in SSc dermal fibroblasts and endothelial cells compared with healthy controls. EZH2 inhibitor DZNep halted fibrosis both in vitro and in vivo. In SSc fibroblasts, DZNep dose-dependently reduced the expression of profibrotic genes and inhibited migratory activity of SSc fibroblasts. We show that epigenetic dysregulation and overexpression of LRRC16A explains EZH2-mediated fibroblast migration in SSc. In endothelial cells, inhibition of EZH2 restored normal angiogenesis in SSc via activating the Notch pathway, specifically by up-regulating the Notch ligand DLL4. Our results demonstrate that overexpression of EZH2 in SSc fibroblasts and endothelial cells is profibrotic and antiangiogenic. Targeting EZH2 or EZH2-regulated genes might be of therapeutic potential in SSc.


Assuntos
Proteína Potenciadora do Homólogo 2 de Zeste/genética , Fibrose/genética , Proteínas dos Microfilamentos/genética , Esclerodermia Difusa/genética , Animais , Bleomicina/toxicidade , Movimento Celular/genética , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Repressão Epigenética/genética , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibrose/induzido quimicamente , Fibrose/patologia , Regulação da Expressão Gênica/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Proteínas de Membrana/genética , Metilação , Camundongos , Neovascularização Fisiológica , Receptores Notch/genética , Transdução de Sinais
5.
FASEB J ; 34(7): 8810-8823, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32445534

RESUMO

The glycoprotein nonmetastatic melanoma protein B (GPNMB, also known as osteoactivin) is highly expressed in many cell types and regulates the homeostasis in various tissues. In different physiological contexts, it functions as a melanosome-associated protein, membrane-bound surface receptor, soluble ligand, or adhesion molecule. Therefore, GPNMB is involved in cell differentiation, migration, inflammation, metabolism, and neuroprotection. Because of its various involvement in different physiological conditions, GPNMB has been implicated in many diseases, including cancer, neurological disorders, and more recently immune-mediated diseases. This review summarizes the regulation and function of GPNMB in normal physiology, and discusses the involvement of GPNMB in disease conditions with a particular focus on its potential role and therapeutic implications in autoimmunity.


Assuntos
Doenças Autoimunes/patologia , Regulação da Expressão Gênica , Imunoterapia/métodos , Glicoproteínas de Membrana/antagonistas & inibidores , Doenças Autoimunes/metabolismo , Doenças Autoimunes/terapia , Humanos , Glicoproteínas de Membrana/metabolismo
6.
Ann Rheum Dis ; 78(4): 519-528, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30674474

RESUMO

OBJECTIVE: We examined genome-wide DNA methylation changes in CD8+ T cells from patients with lupus and controls and investigated the functional relevance of some of these changes in lupus. METHODS: Genome-wide DNA methylation of lupus and age, sex and ethnicity-matched control CD8+ T cells was measured using the Infinium MethylationEPIC arrays. Measurement of relevant cell subsets was performed via flow cytometry. Gene expression was quantified by qPCR. Inhibiting STAT1 and CIITA was performed using fludarabine and CIITA siRNA, respectively. RESULTS: Lupus CD8+ T cells had 188 hypomethylated CpG sites compared with healthy matched controls. Among the most hypomethylated were sites associated with HLA-DRB1. Genes involved in the type-I interferon response, including STAT1, were also found to be hypomethylated. IFNα upregulated HLA-DRB1 expression on lupus but not control CD8+ T cells. Lupus and control CD8+ T cells significantly increased STAT1 mRNA levels after treatment with IFNα. The expression of CIITA, a key interferon/STAT1 dependent MHC-class II regulator, is induced by IFNα in lupus CD8+ T cells, but not healthy controls. CIITA knockdown and STAT1 inhibition experiments revealed that HLA-DRB1 expression in lupus CD8+ T cells is dependent on CIITA and STAT1 signalling. Coincubation of naïve CD4+ T cells with IFNα-treated CD8+ T cells led to CD4+ T cell activation, determined by increased expression of CD69 and cytokine production, in patients with lupus but not in healthy controls. This can be blocked by neutralising antibodies targeting HLA-DR. CONCLUSIONS: Lupus CD8+ T cells are epigenetically primed to respond to type-I interferon. We describe an HLA-DRB1+ CD8+ T cell subset that can be induced by IFNα in patients with lupus. A possible pathogenic role for CD8+ T cells in lupus that is dependent on a high type-I interferon environment and epigenetic priming warrants further characterisation.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Metilação de DNA , Cadeias HLA-DRB1/genética , Lúpus Eritematoso Sistêmico/genética , Fator de Transcrição STAT1/genética , Adulto , Idoso , Linfócitos T CD4-Positivos/imunologia , Estudos de Casos e Controles , Células Cultivadas , Técnicas de Cocultura , Feminino , Regulação da Expressão Gênica/imunologia , Estudo de Associação Genômica Ampla , Humanos , Interferon Tipo I/imunologia , Interferon-alfa/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Ativação Linfocitária/imunologia , Masculino , Pessoa de Meia-Idade , Proteínas Nucleares/genética , RNA Mensageiro/genética , Transativadores/genética , Regulação para Cima/imunologia , Adulto Jovem
7.
Curr Rheumatol Rep ; 21(12): 69, 2019 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-31813068

RESUMO

PURPOSE OF REVIEW: Epigenetics has been implicated in the pathogenesis of systemic sclerosis (SSc). In this review, the involvement of the three epigenetic mechanisms in SSc development and progression-DNA methylation, histone modifications, and non-coding RNAs-will be discussed. RECENT FINDINGS: Alteration in epigenetics was observed in immune cells, dermal fibroblasts, and endothelial cells derived from SSc patients. Genes that are affected include those involved in immune cell function and differentiation, TGFß and Wnt pathways, extracellular matrix accumulation, transcription factors, and angiogenesis. All the studies remain in the pre-clinical stage. Extensive research provides evidence that epigenetic alterations are critical for SSc pathogenesis. Future epigenomic studies will undoubtedly continue to broaden our understanding of disease pathogenesis and clinical heterogeneity. They will also provide the scientific basis for repurposing epigenetic-modifying agents for SSc patients.


Assuntos
Epigenômica , Escleroderma Sistêmico/genética , Escleroderma Sistêmico/terapia , Metilação de DNA , Código das Histonas , Humanos , RNA não Traduzido , Escleroderma Sistêmico/diagnóstico
8.
Clin Immunol ; 192: 68-77, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29678503

RESUMO

Caffeine is a widely consumed pharmacologically active product. We focused on characterizing immunomodulatory effects of caffeine on peripheral blood mononuclear cells. Caffeine at high doses showed a robust downregulatory effect on cytokine activity and genes related to several autoimmune diseases including lupus and rheumatoid arthritis. Dose-dependent validation experiments showed downregulation at the mRNA levels of key inflammation-related genes including STAT1, TNF, IFNG, and PPARG. TNF and PPARG were suppressed even with the lowest caffeine dose tested, which corresponds to the serum concentration of caffeine after administration of one cup of coffee. Cytokine levels of IL-8, MIP-1ß, IL-6, IFN-γ, GM-CSF, TNF, IL-2, IL-4, MCP-1, and IL-10 were decreased significantly with caffeine treatment. Upstream regulator analysis suggests that caffeine inhibits STAT1 signaling, which was confirmed by showing reduced phosphorylated STAT1 after caffeine treatment. Further studies exploring disease-modulating potential of caffeine in autoimmune diseases and further exploring the mechanisms involved are warranted.


Assuntos
Autoimunidade/efeitos dos fármacos , Cafeína/farmacologia , Inflamação/imunologia , Leucócitos Mononucleares/efeitos dos fármacos , Fator de Transcrição STAT1/imunologia , Artrite Reumatoide/genética , Artrite Reumatoide/imunologia , Artrite Reumatoide/metabolismo , Autoimunidade/genética , Autoimunidade/imunologia , Cafeína/administração & dosagem , Estimulantes do Sistema Nervoso Central/farmacologia , Citocinas/genética , Citocinas/imunologia , Citocinas/metabolismo , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Humanos , Inflamação/genética , Inflamação/metabolismo , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , PPAR gama/genética , PPAR gama/imunologia , PPAR gama/metabolismo , Fosforilação/efeitos dos fármacos , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/imunologia
9.
Ann Rheum Dis ; 77(8): 1208-1218, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29760157

RESUMO

OBJECTIVE: Emerging evidence supports a role for epigenetic regulation in the pathogenesis of scleroderma (SSc). We aimed to assess the role of methyl-CpG-binding protein 2 (MeCP2), a key epigenetic regulator, in fibroblast activation and fibrosis in SSc. METHODS: Dermal fibroblasts were isolated from patients with diffuse cutaneous SSc (dcSSc) and from healthy controls. MeCP2 expression was measured by qPCR and western blot. Myofibroblast differentiation was evaluated by gel contraction assay in vitro. Fibroblast proliferation was analysed by ki67 immunofluorescence staining. A wound healing assay in vitro was used to determine fibroblast migration rates. RNA-seq was performed with and without MeCP2 knockdown in dcSSc to identify MeCP2-regulated genes. The expression of MeCP2 and its targets were modulated by siRNA or plasmid. Chromatin immunoprecipitation followed by sequencing (ChIP-seq) using anti-MeCP2 antibody was performed to assess MeCP2 binding sites within MeCP2-regulated genes. RESULTS: Elevated expression of MeCP2 was detected in dcSSc fibroblasts compared with normal fibroblasts. Overexpressing MeCP2 in normal fibroblasts suppressed myofibroblast differentiation, fibroblast proliferation and fibroblast migration. RNA-seq in MeCP2-deficient dcSSc fibroblasts identified MeCP2-regulated genes involved in fibrosis, including PLAU, NID2 and ADA. Plasminogen activator urokinase (PLAU) overexpression in dcSSc fibroblasts reduced myofibroblast differentiation and fibroblast migration, while nidogen-2 (NID2) knockdown promoted myofibroblast differentiation and fibroblast migration. Adenosine deaminase (ADA) depletion in dcSSc fibroblasts inhibited cell migration rates. Taken together, antifibrotic effects of MeCP2 were mediated, at least partly, through modulating PLAU, NID2 and ADA. ChIP-seq further showed that MeCP2 directly binds regulatory sequences in NID2 and PLAU gene loci. CONCLUSIONS: This study demonstrates a novel role for MeCP2 in skin fibrosis and identifies MeCP2-regulated genes associated with fibroblast migration, myofibroblast differentiation and extracellular matrix degradation, which can be potentially targeted for therapy in SSc.


Assuntos
Fibroblastos/metabolismo , Proteína 2 de Ligação a Metil-CpG/genética , Esclerodermia Difusa/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Diferenciação Celular/genética , Movimento Celular/genética , Proliferação de Células/genética , Células Cultivadas , Epigênese Genética , Matriz Extracelular/fisiologia , Feminino , Fibroblastos/patologia , Fibrose , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Masculino , Proteína 2 de Ligação a Metil-CpG/biossíntese , Pessoa de Meia-Idade , Fenótipo , RNA Mensageiro/genética , Esclerodermia Difusa/patologia , Pele/patologia , Cicatrização/genética
10.
Ann Rheum Dis ; 77(11): 1653-1664, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30021804

RESUMO

OBJECTIVE: Skin inflammation and photosensitivity are common in patients with cutaneous lupus erythematosus (CLE) and systemic lupus erythematosus (SLE), yet little is known about the mechanisms that regulate these traits. Here we investigate the role of interferon kappa (IFN-κ) in regulation of type I interferon (IFN) and photosensitive responses and examine its dysregulation in lupus skin. METHODS: mRNA expression of type I IFN genes was analysed from microarray data of CLE lesions and healthy control skin. Similar expression in cultured primary keratinocytes, fibroblasts and endothelial cells was analysed via RNA-seq. IFNK knock-out (KO) keratinocytes were generated using CRISPR/Cas9. Keratinocytes stably overexpressing IFN-κ were created via G418 selection of transfected cells. IFN responses were assessed via phosphorylation of STAT1 and STAT2 and qRT-PCR for IFN-regulated genes. Ultraviolet B-mediated apoptosis was analysed via TUNEL staining. In vivo protein expression was assessed via immunofluorescent staining of normal and CLE lesional skin. RESULTS: IFNK is one of two type I IFNs significantly increased (1.5-fold change, false discovery rate (FDR) q<0.001) in lesional CLE skin. Gene ontology (GO) analysis showed that type I IFN responses were enriched (FDR=6.8×10-04) in keratinocytes not in fibroblast and endothelial cells, and this epithelial-derived IFN-κ is responsible for maintaining baseline type I IFN responses in healthy skin. Increased levels of IFN-κ, such as seen in SLE, amplify and accelerate responsiveness of epithelia to IFN-α and increase keratinocyte sensitivity to UV irradiation. Notably, KO of IFN-κ or inhibition of IFN signalling with baricitinib abrogates UVB-induced apoptosis. CONCLUSION: Collectively, our data identify IFN-κ as a critical IFN in CLE pathology via promotion of enhanced IFN responses and photosensitivity. IFN-κ is a potential novel target for UVB prophylaxis and CLE-directed therapy.


Assuntos
Epiderme/imunologia , Interferon Tipo I/biossíntese , Lúpus Eritematoso Cutâneo/complicações , Transtornos de Fotossensibilidade/etiologia , Adulto , Células Cultivadas , Células Dendríticas/imunologia , Feminino , Humanos , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Queratinócitos/imunologia , Lúpus Eritematoso Cutâneo/imunologia , Masculino , Pessoa de Meia-Idade , Transtornos de Fotossensibilidade/imunologia , RNA Mensageiro/genética , Pele/imunologia , TYK2 Quinase/imunologia , Regulação para Cima/imunologia
11.
J Autoimmun ; 86: 19-28, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29066026

RESUMO

OBJECTIVE: The goal of this study was to comprehensively characterize CD4+CD28+ T cells overexpressing CD11a and KIR genes, and examine the relationship between this T cell subset, genetic risk, and disease activity in lupus. METHODS: The size of the CD4+CD28+KIR+CD11ahi T cell subset was determined by flow cytometry, and total genetic risk for lupus was calculated in 105 female patients using 43 confirmed genetic susceptibility loci. Primary CD4+CD28+KIR+CD11ahi T cells were isolated from lupus patients or were induced from healthy individuals using 5-azacytidine. Genome-wide DNA methylation was analyzed using an array-based approach, and the transcriptome was assessed by RNA sequencing. Transcripts in the CDR3 region were used to assess the TCR repertoire. Chromatin accessibility was determined using ATAC-seq. RESULTS: A total of 31,019 differentially methylated sites were identified in induced KIR+CD11ahi T cells with >99% being hypomethylated. RNA sequencing revealed a clear pro-inflammatory transcriptional profile. TCR repertoire analysis suggests less clonotype diversity in KIR+CD11ahi compared to autologous KIR-CD11alow T cells. Similarly, primary KIR+CD11ahi T cells isolated from lupus patients were hypomethylated and characterized by a pro-inflammatory chromatin structure. We show that the genetic risk for lupus was significantly higher in African-American compared to European-American lupus patients. The demethylated CD4+CD28+KIR+CD11ahi T cell subset size was a better predictor of disease activity in young (age ≤ 40) European-American patients independent of genetic risk. CONCLUSION: CD4+CD28+KIR+CD11ahi T cells are demethylated and characterized by pro-inflammatory epigenetic and transcriptional profiles in lupus. Eliminating these cells or blocking their pro-inflammatory characteristics might present a novel therapeutic approach for lupus.


Assuntos
Negro ou Afro-Americano , Inflamação/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T/imunologia , Antígeno CD11a/metabolismo , Antígenos CD28/metabolismo , Antígenos CD4/metabolismo , Células Cultivadas , Metilação de DNA , Progressão da Doença , Epigênese Genética , Feminino , Perfil Genético , Humanos , Imunofenotipagem , Lúpus Eritematoso Sistêmico/diagnóstico , Lúpus Eritematoso Sistêmico/epidemiologia , Receptores KIR/metabolismo , Risco , Análise de Sequência de RNA , Estados Unidos/epidemiologia
12.
J Autoimmun ; 83: 73-94, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28526340

RESUMO

With unknown etiology, scleroderma (SSc) is a multifaceted disease characterized by immune activation, vascular complications, and excessive fibrosis in internal organs. Genetic studies, including candidate gene association studies, genome-wide association studies, and whole-exome sequencing have supported the notion that while genetic susceptibility to SSc appears to be modest, SSc patients are genetically predisposed to this disease. The strongest genetic association for SSc lies within the MHC region, with loci in HLA-DRB1, HLA-DQB1, HLA-DPB1, and HLA-DOA1 being the most replicated. The non-HLA genes associated with SSc are involved in various functions, with the most robust associations including genes for B and T cell activation and innate immunity. Other pathways include genes involved in extracellular matrix deposition, cytokines, and autophagy. Among these genes, IRF5, STAT4, and CD247 were replicated most frequently while SNPs rs35677470 in DNASE1L3, rs5029939 in TNFAIP3, and rs7574685 in STAT4 have the strongest associations with SSc. In addition to genetic predisposition, it became clear that environmental factors and epigenetic influences also contribute to the development of SSc. Epigenetics, which refers to studies that focus on heritable phenotypes resulting from changes in chromatin structure without affecting the DNA sequence, is one of the most rapidly expanding fields in biomedical research. Indeed extensive epigenetic changes have been described in SSc. Alteration in enzymes and mediators involved in DNA methylation and histone modification, as well as dysregulated non-coding RNA levels all contribute to fibrosis, immune dysregulation, and impaired angiogenesis in this disease. Genes that are affected by epigenetic dysregulation include ones involved in autoimmunity, T cell function and regulation, TGFß pathway, Wnt pathway, extracellular matrix, and transcription factors governing fibrosis and angiogenesis. In this review, we provide a comprehensive overview of the current findings of SSc genetic susceptibility, followed by an extensive description and a systematic review of epigenetic research that has been carried out to date in SSc. We also summarize the therapeutic potential of drugs that affect epigenetic mechanisms, and outline the future prospective of genomics and epigenomics research in SSc.


Assuntos
Antígenos HLA/genética , Fatores Reguladores de Interferon/genética , Fator de Transcrição STAT4/genética , Escleroderma Sistêmico/genética , Epigenômica , Interação Gene-Ambiente , Predisposição Genética para Doença , Genômica , Humanos , Polimorfismo Genético , Escleroderma Sistêmico/imunologia , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/metabolismo
13.
Rheumatology (Oxford) ; 56(11): 1970-1981, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28968684

RESUMO

Objectives: SSc is a devastating disease that results in fibrosis of the skin and other organs. Fibroblasts are a key driver of the fibrotic process through deposition of extracellular matrix. The mechanisms by which fibroblasts are induced to become pro-fibrotic remain unclear. Thus, we examined the ability of SSc keratinocytes to promote fibroblast activation and the source of this effect. Methods: Keratinocytes were isolated from skin biopsies of 9 lcSSc, 10 dcSSc and 13 control patients. Conditioned media was saved from the cultures. Normal fresh primary fibroblasts were exposed to healthy control and SSc keratinocyte conditioned media in the presence or absence of neutralizing antibodies for TGF-ß. Gene expression was assessed by microarrays and real-time PCR. Immunocytochemistry was performed for α-smooth muscle actin (α-SMA), collagen type 1 (COL1A1) and CCL5 expression. Results: SSc keratinocyte conditioned media promoted fibroblast activation, characterized by increased α-SMA and COL1A1 mRNA and protein expression. This effect was independent of TGF-ß. Microarray analysis identified upregulation of nuclear factor κB (NF-κB) and downregulation of peroxisome proliferator-activated receptor γ (PPAR-γ) pathways in both SSc subtypes. Scleroderma keratinocytes exhibited increased expression of NF-κB-regulated cytokines and chemokines and lesional skin staining confirmed upregulation of CCL5 in basal keratinocytes. Conclusion: Scleroderma keratinocytes promote the activation of fibroblasts in a TGF-ß-independent manner and demonstrate an imbalance in NF-κB1 and PPAR-γ expression leading to increased cytokine and CCL5 production. Further study of keratinocyte mediators of fibrosis, including CCL5, may provide novel targets for skin fibrosis therapy.


Assuntos
Diferenciação Celular , Fibroblastos/citologia , Queratinócitos/citologia , Escleroderma Sistêmico/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Actinas/metabolismo , Adulto , Idoso , Células Cultivadas , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Meios de Cultivo Condicionados , Regulação para Baixo , Feminino , Fibroblastos/metabolismo , Fibrose , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Queratinócitos/metabolismo , Masculino , Pessoa de Meia-Idade , NF-kappa B/metabolismo , PPAR gama/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Esclerodermia Difusa , Esclerodermia Localizada , Escleroderma Sistêmico/genética , Escleroderma Sistêmico/patologia , Pele/patologia , Regulação para Cima
14.
Bioorg Med Chem Lett ; 27(8): 1744-1749, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28285914

RESUMO

We recently reported the development of a novel inhibitor of Rho-mediated gene transcription (1, CCG-203971) that is efficacious in multiple animal models of acute fibrosis, including scleroderma, when given intraperitoneally. The modest in vivo potency and poor pharmacokinetics (PK) of this lead, however, make it unsuitable for long term efficacy studies. We therefore undertook a systematic medicinal chemistry effort to improve both the metabolic stability and the solubility of 1, resulting in the identification of two analogs achieving over 10-fold increases in plasma exposures in mice. We subsequently showed that one of these analogs (8f, CCG-232601) could inhibit the development of bleomycin-induced dermal fibrosis in mice when administered orally at 50mg/kg, an effect that was comparable to what we had observed earlier with 1 at a 4-fold higher IP dose.


Assuntos
Ácidos Nipecóticos/farmacocinética , Ácidos Nipecóticos/uso terapêutico , Fator Rho/antagonistas & inibidores , Escleroderma Sistêmico/tratamento farmacológico , Pele/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos , Administração Oral , Animais , Modelos Animais de Doenças , Fibrose , Células HEK293 , Humanos , Camundongos , Ácidos Nipecóticos/administração & dosagem , Ácidos Nipecóticos/química , Fator Rho/metabolismo , Escleroderma Sistêmico/genética , Escleroderma Sistêmico/metabolismo , Escleroderma Sistêmico/patologia , Elemento de Resposta Sérica/efeitos dos fármacos , Pele/metabolismo , Pele/patologia , Transativadores/antagonistas & inibidores , Transativadores/metabolismo
15.
Rheumatology (Oxford) ; 55(4): 745-54, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26705326

RESUMO

OBJECTIVES: Angiogenesis plays a critical role in SSc (scleroderma). The aim of this study was to examine the expression of growth-regulated protein-γ (Gro-γ/CXCL3), granulocyte chemotactic protein 2 (GCP-2/CXCL6) and their receptor CXCR2 in endothelial cells (ECs) isolated from SSc skin and determine whether these cells mount an angiogenic response towards pro-angiogenic chemokines. The downstream signalling pathways as well as the pro-angiogenic transcription factor inhibitor of DNA-binding protein 1 (Id-1) were also examined. METHODS: Skin biopsies were obtained from patients with dcSSc. ECs were isolated via magnetic positive selection. Angiogenesis was measured by EC chemotaxis assay. RESULTS: Gro-γ/CXCL3 and GCP-2/CXCL6 were minimally expressed in both skin types but elevated in SSc serum. Pro-angiogenic chemokine mRNA was greater in SSc ECs than in normal ECs. SSc ECs did not migrate to vascular endothelial growth factor (VEGF), Gro-γ/CXCL3, GCP-2/CXCL6 or CXCL16. The signalling pathways stimulated by these chemokines were also dysregulated. Id-1 mRNA in SSc ECs was lower compared with normal ECs, and overexpression of Id-1 in SSc ECs increased their ability to migrate towards VEGF and CXCL16. CONCLUSION: Our results show that SSc ECs are unable to respond to pro-angiogenic chemokines despite their increased expression in serum and ECs. This might be due to the differences in the signalling pathways activated by these chemokines in normal vs SSc ECs. In addition, the lower expression of Id-1 also decreases the angiogenic response. The inability of pro-angiogenic chemokines to promote EC migration provides an additional mechanism for the impaired angiogenesis that characterizes SSc.


Assuntos
Quimiocinas/fisiologia , Endotélio Vascular/patologia , Neovascularização Patológica/patologia , Escleroderma Sistêmico/patologia , Pele/irrigação sanguínea , Indutores da Angiogênese/farmacologia , Estudos de Casos e Controles , Células Cultivadas , Quimiocinas/biossíntese , Quimiocinas/farmacologia , Quimiotaxia/efeitos dos fármacos , Quimiotaxia/fisiologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Feminino , Humanos , Proteína 1 Inibidora de Diferenciação/fisiologia , Masculino , Pessoa de Meia-Idade , Neovascularização Patológica/metabolismo , Receptores de Interleucina-8B/metabolismo , Escleroderma Sistêmico/metabolismo , Transdução de Sinais/fisiologia , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/farmacologia
16.
Ann Rheum Dis ; 74(8): 1612-20, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24812288

RESUMO

BACKGROUND: The aetiology of systemic sclerosis (SSc) is not clear, but there is an emerging evidence of gene-specific epigenetic dysregulation in the pathogenesis of SSc. METHODS: We performed a genome-wide DNA methylation study in dermal fibroblasts in six diffuse cutaneous SSc (dSSc) patients, six limited cutaneous SSc (lSSc) patients compared with 12 age-matched, sex-matched and ethnicity-matched healthy controls. Cytosine methylation was quantified in more than 485 000 methylation sites across the genome. Differentially methylated CpG sites between patients and controls with a fold difference ≥1.2 were identified. Quantitative real-time RT-PCR was performed to assess correlation between DNA methylation changes and gene expression levels. RESULTS: We identified 2710 and 1021 differentially methylated CpG sites in dSSc and lSSc, respectively. Of the differentially methylated sites, 61% in dSSc and 90% in lSSc were hypomethylated. There were only 203 CpG sites differentially methylated in both dSSc and lSSc, representing 118 hypomethylated and 6 hypermethylated genes. Common hypomethylated genes include ITGA9, encoding an α integrin. Other relevant genes such as ADAM12, COL23A1, COL4A2 and MYO1E, and transcription factors genes RUNX1, RUNX2 and RUNX3 were also hypomethylated in both dSSc and lSSc. Pathway analysis of differentially methylated genes in both dSSc and lSSc revealed enrichment of genes involved in extracellular matrix-receptor interaction and focal adhesion. We demonstrate significant correlation between DNA methylation status and gene expression in the majority of genes evaluated. CONCLUSIONS: Our data highlight common and subset-specific aberrancies in dSSc and lSSc fibroblasts at the epigenomic levels and identify novel candidate genes in SSc.


Assuntos
Metilação de DNA , Fibroblastos/metabolismo , Esclerodermia Difusa/metabolismo , Esclerodermia Limitada/metabolismo , Pele/citologia , Adulto , Idoso , Epigenômica , Feminino , Estudo de Associação Genômica Ampla , Humanos , Masculino , Pessoa de Meia-Idade
17.
Am J Physiol Cell Physiol ; 307(1): C2-13, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24740541

RESUMO

Tissue fibrosis occurs with excessive extracellular matrix deposition from myofibroblasts, resulting in tissue scarring and inflammation. It is driven by multiple mediators, such as the G protein-coupled receptor ligands lysophosphatidic acid and endothelin, as well as signaling by transforming growth factor-ß, connective tissue growth factor, and integrins. Fibrosis contributes to 45% of deaths in the developed world. As current therapeutic options for tissue fibrosis are limited and organ transplantation is the only effective treatment for end-stage disease, there is an imminent need for efficacious antifibrotic therapies. This review discusses the various molecular pathways involved in fibrosis. It highlights the Rho GTPase signaling pathway and its downstream gene transcription output through myocardin-related transcription factor and serum response factor as a convergence point for targeting this complex set of diseases.


Assuntos
Doenças do Tecido Conjuntivo/enzimologia , Desenho de Fármacos , Miofibroblastos/enzimologia , Transcrição Gênica , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Doenças do Tecido Conjuntivo/tratamento farmacológico , Doenças do Tecido Conjuntivo/genética , Doenças do Tecido Conjuntivo/patologia , Inibidores Enzimáticos/farmacologia , Fibrose , Humanos , Terapia de Alvo Molecular , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/patologia , Fator de Resposta Sérica/genética , Fator de Resposta Sérica/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos
18.
J Pharmacol Exp Ther ; 349(3): 480-6, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24706986

RESUMO

Systemic sclerosis (SSc), or scleroderma, similar to many fibrotic disorders, lacks effective therapies. Current trials focus on anti-inflammatory drugs or targeted approaches aimed at one of the many receptor mechanisms initiating fibrosis. In light of evidence that a myocardin-related transcription factor (MRTF)-and serum response factor (SRF)-regulated gene transcriptional program induced by Rho GTPases is essential for myofibroblast activation, we explored the hypothesis that inhibitors of this pathway may represent novel antifibrotics. MRTF/SRF-regulated genes show spontaneously increased expression in primary dermal fibroblasts from patients with diffuse cutaneous SSc. A novel small-molecule inhibitor of MRTF/SRF-regulated transcription (CCG-203971) inhibits expression of connective tissue growth factor (CTGF), α-smooth muscle actin (α-SMA), and collagen 1 (COL1A2) in both SSc fibroblasts and in lysophosphatidic acid (LPA)-and transforming growth factor ß (TGFß)-stimulated fibroblasts. In vivo treatment with CCG-203971 also prevented bleomycin-induced skin thickening and collagen deposition. Thus, targeting the MRTF/SRF gene transcription pathway could provide an efficacious new approach to therapy for SSc and other fibrotic disorders.


Assuntos
Proteínas de Ligação a DNA/genética , Miofibroblastos/efeitos dos fármacos , Ácidos Nipecóticos/uso terapêutico , Proteínas de Fusão Oncogênica/genética , Escleroderma Sistêmico/tratamento farmacológico , Fator de Resposta Sérica/genética , Transcrição Gênica/efeitos dos fármacos , Animais , Técnicas de Cultura de Células , Proliferação de Células/efeitos dos fármacos , Colágeno Tipo I/genética , Fator de Crescimento do Tecido Conjuntivo/genética , Modelos Animais de Doenças , Feminino , Humanos , Lisofosfolipídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Células NIH 3T3 , Ácidos Nipecóticos/administração & dosagem , Ácidos Nipecóticos/farmacologia , Escleroderma Sistêmico/genética , Escleroderma Sistêmico/patologia , Transativadores , Fator de Crescimento Transformador beta/farmacologia
19.
Trends Mol Med ; 2024 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-38890028

RESUMO

Cellular senescence is a key hallmark of aging. It has now emerged as a key mediator in normal tissue turnover and is associated with a variety of age-related diseases, including organ-specific fibrosis and systemic sclerosis (SSc). This review discusses the recent evidence of the role of senescence in tissue fibrosis, with an emphasis on SSc, a systemic autoimmune rheumatic disease. We discuss the physiological role of these cells, their role in fibrosis, and that targeting these cells specifically could be a new therapeutic avenue in fibrotic disease. We argue that targeting senescent cells, with senolytics or senomorphs, is a viable therapeutic target in fibrotic diseases which remain largely intractable.

20.
Nat Commun ; 15(1): 210, 2024 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-38172207

RESUMO

Systemic sclerosis (SSc) is a devastating autoimmune disease characterized by excessive production and accumulation of extracellular matrix, leading to fibrosis of skin and other internal organs. However, the main cellular participants in SSc skin fibrosis remain incompletely understood. Here using differentiation trajectories at a single cell level, we demonstrate a dual source of extracellular matrix deposition in SSc skin from both myofibroblasts and endothelial-to-mesenchymal-transitioning cells (EndoMT). We further define a central role of Hippo pathway effectors in differentiation and homeostasis of myofibroblast and EndoMT, respectively, and show that myofibroblasts and EndoMTs function as central communication hubs that drive key pro-fibrotic signaling pathways in SSc. Together, our data help characterize myofibroblast differentiation and EndoMT phenotypes in SSc skin, and hint that modulation of the Hippo pathway may contribute in reversing the pro-fibrotic phenotypes in myofibroblasts and EndoMTs.


Assuntos
Via de Sinalização Hippo , Escleroderma Sistêmico , Humanos , Fibrose , Escleroderma Sistêmico/patologia , Miofibroblastos/metabolismo , Células Endoteliais/metabolismo , Pele/patologia , Fibroblastos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA