Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Eur Rev Med Pharmacol Sci ; 25(1): 301-312, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33506919

RESUMO

OBJECTIVE: Coronary artery disease (CAD) is the main cause of mortality worldwide. How stable coronary artery disease (SCAD) progresses to acute myocardial infarction (AMI) is not known. This study was aimed to explore the differentially expressed genes (DEGs) and pathways involved in the progression of SCAD to AMI. MATERIALS AND METHODS: Publicly available gene-expression profiles (GSE71226, GSE97320, GSE66360) were downloaded from the Gene Expression Omnibus (GEO) database and integrated to identify DEGs. The GSE59867 dataset was further used to verify the result of screened DEGs. Functional-enrichment analyses, protein-protein interaction network, microRNA-transcription factor (TF)-mRNA regulatory network, and drug-gene network were visualized. RESULTS: Sixty common DEGs (CDEGs) were screened between the SCAD-Control group and AMI-Control group in the integrated dataset. Four upregulated DEGs were selected from GSE59867. Twenty hub genes were discovered, and three significant modules were constructed in the PPI network. The intersection of functional and pathway-enrichment analyses of 60 CDEGs and the module DEGs indicated that they were mainly involved in "inflammatory response", "immune response", and "cytokine-cytokine receptor interaction". A miRNA-TF-mRNA regulatory network comprised 87 miRNAs, 16 upregulated target DEGs and 7 TFs. CONCLUSIONS: We identified several important genes and miRNAs involved in the progression of SCAD to AMI: platelet activating factor receptor (PTAFR), aquaoporin-9 (AQP9), toll-like receptor-4 (TLR4), human constitutive androstane receptor-3 (HCAR3), leucine-rich-α2 glycoprotein-1 (LRG1), mothers Against Decapentaplegic Homolog 4 (SMAD4) and miRNA-149-5p, miRNA-6778-3p, and miRNA-520a-3p. Inflammation and the immune response had important roles in the progression from SCAD to AMI.


Assuntos
Biologia Computacional , Doença da Artéria Coronariana/metabolismo , Infarto do Miocárdio/metabolismo , Doença Aguda , Aquaporinas/genética , Aquaporinas/imunologia , Doença da Artéria Coronariana/genética , Doença da Artéria Coronariana/patologia , Glicoproteínas/genética , Glicoproteínas/imunologia , Humanos , MicroRNAs/genética , MicroRNAs/imunologia , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Glicoproteínas da Membrana de Plaquetas/genética , Glicoproteínas da Membrana de Plaquetas/metabolismo , Mapas de Interação de Proteínas , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/imunologia , Receptores Acoplados a Proteínas G/metabolismo , Receptores Nicotínicos/genética , Receptores Nicotínicos/imunologia , Proteína Smad4/genética , Proteína Smad4/imunologia , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia
2.
Aging (Albany NY) ; 13(3): 3386-3404, 2021 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-33428605

RESUMO

In this study, we investigated the role of tumor-associated macrophages (TAMs) in the progression of pancreatic ductal adenocarcinoma (PDAC). PDAC patients with higher levels of CD68+ TAMs exhibited shorter overall survival. In Transwell assays, PDAC cells incubated with TAMs or conditioned media from TAM cells (TAM-CM) showed higher migration and invasion rates than controls. PET/CT scan analysis of orthotopic PDAC model mice revealed greater primary tumor growth and liver metastasis in the TAM-CM treatment group than the controls. H&E staining of liver tissues showed significantly higher numbers of metastatic nodules in the TAM-CM treatment group. Heat inactivation of TAM-CM significantly reduced Transwell migration by PDAC cells, suggesting the involvement of one or more secreted proteins in PDAC progression. Transcriptome sequencing analysis of PDAC cells treated with TAM-CM revealed significant enrichment of transforming growth factor-ß (TGF-ß) signaling pathway genes. Western blot and qRT-PCR analysis showed that TAM-CM enhanced PDAC migration cells by inducing epithelial-to-mesenchymal transition through the TGF-ß-Smad2/3/4-Snail signaling axis. The pro-tumorigenic effects of TAMs or TAM-CM were abolished by TGF-ß signaling pathway inhibitors and neutralizing TGF-ß antibody. These results demonstrate that TAMs promote PDAC progression through the TGF-ß signaling pathway.


Assuntos
Carcinoma Ductal Pancreático/imunologia , Transição Epitelial-Mesenquimal/imunologia , Neoplasias Pancreáticas/imunologia , Macrófagos Associados a Tumor/imunologia , Animais , Carcinoma Ductal Pancreático/secundário , Linhagem Celular Tumoral , Movimento Celular , Meios de Cultivo Condicionados , Progressão da Doença , Perfilação da Expressão Gênica , Humanos , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/secundário , Camundongos , Invasividade Neoplásica , Transplante de Neoplasias , Neoplasias Pancreáticas/patologia , Prognóstico , Proteína Smad2/imunologia , Proteína Smad3/imunologia , Proteína Smad4/imunologia , Fatores de Transcrição da Família Snail/imunologia , Células THP-1 , Fator de Crescimento Transformador beta/imunologia
3.
Int J Mol Sci ; 21(19)2020 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-32998457

RESUMO

MicroRNAs regulate gene expression of transcriptional factors, which influence Th17/Treg (regulatory T cells) balance, establishing the molecular mechanism of genetic and epigenetic regulation of Treg and Th17 cells is crucial for understanding rheumatoid arthritis (RA) pathogenesis. The study goal was to understand the potential impact of the selected microRNAs expression profiles on Treg/Th17 cells frequency, RA phenotype, the expression profile of selected microRNAs, and their correlation with the expression profiles of selected transcriptional factors: SOCS1, SMAD3, SMAD4, STAT3, STAT5 in RA; we used osteoarthritis (OA) and healthy controls (HCs) as controls. The study was conducted on 14 RA and 11 OA patients, and 15 HCs. Treg/Th17 frequency was established by flow cytometry. Gene expression analysis was estimated by qPCR. We noticed correlations in RA Th17 cells between miR-26 and SMAD3, STAT3, SOCS1; and miR-155 and STAT3-and in RA Treg cells between miR-26 and SOCS1; miR-31, -155 and SMAD3; and miR-155 and SMAD4. In RA Tregs, we found a negative correlation between miR-26, -126 and STAT5a. The expression level of miR-31 in Th17 cells from RA patients with DAS28 ≤ 5.1 is higher and that for miR-24 is greater in Tregs from patients with DAS28 > 5.1. MiR-146a in Tregs is higher in rheumatoid factor (RF) positive RA patients.


Assuntos
Artrite Reumatoide/genética , MicroRNAs/genética , Osteoartrite/genética , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Artrite Reumatoide/imunologia , Artrite Reumatoide/patologia , Estudos de Casos e Controles , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Masculino , MicroRNAs/imunologia , Pessoa de Meia-Idade , Osteoartrite/imunologia , Osteoartrite/patologia , Fenótipo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/imunologia , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/imunologia , Índice de Gravidade de Doença , Proteína Smad3/genética , Proteína Smad3/imunologia , Proteína Smad4/genética , Proteína Smad4/imunologia , Proteína 1 Supressora da Sinalização de Citocina/genética , Proteína 1 Supressora da Sinalização de Citocina/imunologia , Linfócitos T Reguladores/patologia , Células Th17/patologia
4.
Mol Immunol ; 114: 260-268, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31398665

RESUMO

Th17 cells are essential for the pathogenesis of inflammatory and autoimmune diseases. In the presence of TGF-ß, the differentiation of Th17 cells can be induced by inflammatory cytokines, especially IL-6, which is mainly produced by antigen presenting cells (APCs); or IL-21, which is derived from T cells. IL-21 is required for IL-6-induced Th17 cell differentiation. However, the key regulators and underlying mechanisms for IL-21-induced Th17 differentiation is still elusive. Here we show that SMAD4 is a key regulator in IL-21-induced Th17 differentiation. SMAD4 deficient naïve T cells can differentiate into Th17 cells in the absence of TGF-ß signaling, and these Th17 cells are pathogenic during EAE. SMAD4 represses Rorc mRNA transcription to constrain IL-21-induced Th17 differentiation in the absence of TGF-ß signaling. While in the presence of TGF-ß, SMAD4 losses its suppressive ability due to the degradation of SKI. Mutation of Y429A or A432E on SMAD4 disrupts the interaction of SKI from SMAD4 and eliminates SMAD4 mediated suppression of Th17 differentiation. SMAD4 is indispensable for SKI binding to Rorc promoter region to regulate Th17 differentiation. Moreover, activin can induce Th17 differentiation in combination with IL-21, and the process is also subjected to the control of SKI and SMAD4. This study therefore elucidates critical mechanism for IL-21-induced Th17 differentiation to indicate SKI and SMAD4 as potential therapeutic targets for treating autoimmune diseases.


Assuntos
Diferenciação Celular/imunologia , Interleucinas/imunologia , Proteína Smad4/imunologia , Células Th17/imunologia , Animais , Doenças Autoimunes/imunologia , Citocinas/imunologia , Inflamação/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/imunologia , Fator de Crescimento Transformador beta/imunologia
5.
Front Immunol ; 10: 904, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31118932

RESUMO

We characterized the NK cell phenotype and function in three family members with Hereditary Hemorrhagic Telangiectasia (HHT) due to heterozygous SMAD4 mutations. Loss-of-function mutation in this gene did not induce developmental effects to alter CD56bright or CD56dim NK cell subset proportions in peripheral blood; and did not result in major differences in either their IL-15-induced proliferation, or their cytokine secretion response to TGF-ß1. These data suggest that SMAD4 plays a redundant role in downstream TGF-ß signaling in NK cells.


Assuntos
Células Matadoras Naturais/imunologia , Proteína Smad4/imunologia , Telangiectasia Hemorrágica Hereditária/imunologia , Fator de Crescimento Transformador beta/imunologia , Idoso , Feminino , Humanos , Mutação com Perda de Função , Masculino , Pessoa de Meia-Idade , Proteína Smad4/genética , Telangiectasia Hemorrágica Hereditária/genética
6.
Clin Cancer Res ; 25(6): 1948-1956, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30587545

RESUMO

PURPOSE: SMAD4 has shown promise in identifying patients with colorectal cancer at high risk of recurrence or death.Experimental Design: A discovery cohort and independent validation cohort were classified by SMAD4 status. SMAD4 status and immune infiltrate measurements were tested for association with recurrence-free survival (RFS). Patient-derived xenografts from SMAD4-deficient and SMAD4-retained tumors were used to examine chemoresistance. RESULTS: The discovery cohort consisted of 364 patients with stage I-IV colorectal cancer. Median age at diagnosis was 53 years. The cohort consisted of 61% left-sided tumors and 62% stage II/III patients. Median follow-up was 5.4 years (interquartile range, 2.3-8.2). SMAD4 loss, noted in 13% of tumors, was associated with higher tumor and nodal stage, adjuvant therapy use, fewer tumor-infiltrating lymphocytes (TIL), and lower peritumoral lymphocyte aggregate (PLA) scores (all P < 0.04). SMAD4 loss was associated with worse RFS (P = 0.02). When stratified by SMAD4 and immune infiltrate status, patients with SMAD4 loss and low TIL or PLA had worse RFS (P = 0.002 and P = 0.006, respectively). Among patients receiving 5-fluorouracil (5-FU)-based systemic chemotherapy, those with SMAD4 loss had a median RFS of 3.8 years compared with 13 years for patients with SMAD4 retained. In xenografted mice, the SMAD4-lost tumors displayed resistance to 5-FU. An independent cohort replicated our findings, in particular, the association of SMAD4 loss with decreased immune infiltrate, as well as worse disease-specific survival. CONCLUSIONS: Our data show SMAD4 loss correlates with worse clinical outcome, resistance to chemotherapy, and decreased immune infiltrate, supporting its use as a prognostic marker in patients with colorectal cancer.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Biomarcadores Tumorais/deficiência , Neoplasias Colorretais/patologia , Recidiva Local de Neoplasia/diagnóstico , Proteína Smad4/deficiência , Adulto , Idoso , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Biomarcadores Tumorais/imunologia , Quimioterapia Adjuvante/métodos , Colo/patologia , Colo/cirurgia , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/terapia , Intervalo Livre de Doença , Resistencia a Medicamentos Antineoplásicos/imunologia , Feminino , Fluoruracila/farmacologia , Fluoruracila/uso terapêutico , Seguimentos , Humanos , Linfócitos do Interstício Tumoral/imunologia , Masculino , Camundongos , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/imunologia , Recidiva Local de Neoplasia/patologia , Recidiva Local de Neoplasia/prevenção & controle , Prognóstico , Estudos Prospectivos , Reto/patologia , Reto/cirurgia , Proteína Smad4/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Nutr Cancer ; 70(7): 1075-1087, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30273070

RESUMO

Pancreatic cancer (PC) patients have poor prognosis and survival rate. Gemcitabine, the drug of choice has a dismal 15% response rate. Earlier, we reported that Garcinol alone and in combination with gemcitabine showed a dose-dependent favorable response on PC cell lines. This study probes the in vivo effects of dietary Garcinol on PC progression in transgenic PC mice (KPC; K-ras and p53 conditional mutant). KPC male mice were divided into: KC- Control diet; KGr-0.05% Garcinol diet; KGm-Gemcitabine injected; KGG - Garcinol diet + Gemcitabine injected groups. Changes in tumor progression, toxicity, or cell morphology were monitored by magnetic resonance imaging, Fore-stomach, and blood smear, respectively. Pancreatic Intraepithelial Neoplasia (mPanIN) grading with hematoxylin and eosin (H&E) staining was conducted on pancreas and validated by immunohistochemistry. The KGr group showed improved survival, no observable toxicity with marked reduction in papilloma formation in the fore-stomach, and a higher ratio of NK and NKT cells compared to Non-NK lymphocytes. Additionally, the KGr, KGm, and KGG groups showed reduction in tumor volumes and reduced number of advanced mouse PanIN3. Dietary Garcinol alone and in combination with gemcitabine retarded the progression of PC in transgenic PC mice, arresting the cancer in the earlier stages, improving prognosis and survival.


Assuntos
Neoplasias Pancreáticas/dietoterapia , Terpenos/farmacologia , Animais , Antineoplásicos Fitogênicos/efeitos adversos , Antineoplásicos Fitogênicos/farmacologia , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Suplementos Nutricionais , Genes p53 , Genes ras , Humanos , Imageamento por Ressonância Magnética , Masculino , Camundongos Transgênicos , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Neoplasias Pancreáticas/diagnóstico por imagem , Neoplasias Pancreáticas/tratamento farmacológico , Subunidade beta da Proteína Ligante de Cálcio S100/imunologia , Proteína Smad4/imunologia , Taxa de Sobrevida , Terpenos/efeitos adversos , Gencitabina
8.
J Clin Invest ; 128(11): 5123-5136, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30183689

RESUMO

SMAD4 is the only common SMAD in TGF-ß signaling that usually impedes immune cell activation in the tumor microenvironment. However, we demonstrated here that selective deletion of Smad4 in NK cells actually led to dramatically reduced tumor cell rejection and augmented tumor cell metastases, reduced murine CMV clearance, as well as impeded NK cell homeostasis and maturation. This was associated with a downregulation of granzyme B (Gzmb), Kit, and Prdm1 in Smad4-deficient NK cells. We further unveiled the mechanism by which SMAD4 promotes Gzmb expression. Gzmb was identified as a direct target of a transcriptional complex formed by SMAD4 and JUNB. A JUNB binding site distinct from that for SMAD4 in the proximal Gzmb promoter was required for transcriptional activation by the SMAD4-JUNB complex. In a Tgfbr2 and Smad4 NK cell-specific double-conditional KO model, SMAD4-mediated events were found to be independent of canonical TGF-ß signaling. Our study identifies and mechanistically characterizes unusual functions and pathways for SMAD4 in governing innate immune responses to cancer and viral infection, as well as NK cell development.


Assuntos
Imunidade Inata , Células Matadoras Naturais/imunologia , Melanoma Experimental/imunologia , Proteínas de Neoplasias/imunologia , Transdução de Sinais/imunologia , Proteína Smad4/imunologia , Fator de Crescimento Transformador beta/imunologia , Animais , Regulação Neoplásica da Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/imunologia , Granzimas/genética , Granzimas/imunologia , Células Matadoras Naturais/patologia , Melanoma Experimental/genética , Melanoma Experimental/patologia , Camundongos , Camundongos Knockout , Proteínas de Neoplasias/genética , Receptor do Fator de Crescimento Transformador beta Tipo II/genética , Receptor do Fator de Crescimento Transformador beta Tipo II/imunologia , Transdução de Sinais/genética , Proteína Smad4/genética , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia , Fator de Crescimento Transformador beta/genética
9.
Cell Mol Gastroenterol Hepatol ; 6(3): 257-276, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30109253

RESUMO

Background & Aims: Chronic inflammation is a predisposing condition for colorectal cancer. Many studies to date have focused on proinflammatory signaling pathways in the colon. Understanding the mechanisms that suppress inflammation, particularly in epithelial cells, is critical for developing therapeutic interventions. Here, we explored the roles of transforming growth factor ß (TGFß) family signaling through SMAD4 in colonic epithelial cells. Methods: The Smad4 gene was deleted specifically in adult murine intestinal epithelium. Colitis was induced by 3 rounds of dextran sodium sulfate in drinking water, after which mice were observed for up to 3 months. Nontransformed mouse colonocyte cell lines and colonoid cultures and human colorectal cancer cell lines were analyzed for responses to TGFß1 and bone morphogenetic protein 2. Results: Dextran sodium sulfate treatment was sufficient to drive carcinogenesis in mice lacking colonic Smad4 expression, with resulting tumors bearing striking resemblance to human colitis-associated carcinoma. Loss of SMAD4 protein was observed in 48% of human colitis-associated carcinoma samples as compared with 19% of sporadic colorectal carcinomas. Loss of Smad4 increased the expression of inflammatory mediators within nontransformed mouse colon epithelial cells in vivo. In vitro analysis of mouse and human colonic epithelial cell lines and organoids indicated that much of this regulation was cell autonomous. Furthermore, TGFß signaling inhibited the epithelial inflammatory response to proinflammatory cytokines. Conclusions: TGFß suppresses the expression of proinflammatory genes in the colon epithelium, and loss of its downstream mediator, SMAD4, is sufficient to initiate inflammation-driven colon cancer. Transcript profiling: GSE100082.


Assuntos
Carcinoma/imunologia , Colite/imunologia , Neoplasias Colorretais/imunologia , Inflamação/imunologia , Proteína Smad4/imunologia , Animais , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/metabolismo , Carcinoma/etiologia , Carcinoma/patologia , Linhagem Celular , Linhagem Celular Tumoral , Colite/induzido quimicamente , Colite/complicações , Neoplasias Colorretais/etiologia , Neoplasias Colorretais/patologia , Sulfato de Dextrana/farmacologia , Humanos , Inflamação/induzido quimicamente , Inflamação/complicações , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteína Smad4/genética , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo
10.
Redox Biol ; 18: 104-113, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30007886

RESUMO

Elevated levels of mitochondrial reactive oxygen species (ROS) can lead to the development of airway inflammation. In this study, we investigated the role of Aspergillus proteases-which contribute to the pathogenesis of Aspergillus-induced diseases such as allergic bronchopulmonary aspergillosis, hypersensitivity pneumonitis, and atopic asthma-and their mechanisms of action in airway inflammation using primary human bronchial epithelial cells, and evaluated the inflammatory responses mediated by mitochondrial ROS. We found that Aspergillus proteases regulated the expression of multifunctional inflammatory cytokines such as interleukin (IL)- 1ß, - 6, and - 8, and transforming growth factor (TGF)-ß, which stimulated cytokine production and chemokines involved in leukocyte migration and activated an inflammatory cascade. Expression of these factors and activator protein (AP)- 1 were decreased by treatment with the mitochondrial ROS scavenger Mito-TEMPO, suggesting that mitochondria are important sources of ROS in the context of inflammatory response by Aspergillus protease. The regulation of mitochondrial ROS influenced the production of proinflammatory mediators by preventing mitochondrial ROS-induced AP-1 activation in airway epithelial cells. In addition, Aspergillus protease-mediated mitochondrial ROS production was associated with downregulation of uncoupling protein (UCP)- 2 expression by TGF-ß-SMAD4 signaling, which may play a regulatory role in mitochondrial ROS formation during fungal protease-mediated epithelial inflammation. This improved understanding of the allergenic fungal protease-induced inflammatory mechanism in the bronchial epithelium will help in developing intervention strategies for the regulation of inflammatory response in allergic airway diseases.


Assuntos
Aspergillus oryzae/imunologia , Brônquios/microbiologia , Proteínas Fúngicas/imunologia , Inflamação/imunologia , Peptídeo Hidrolases/imunologia , Proteína Smad4/imunologia , Fator de Crescimento Transformador beta/imunologia , Proteína Desacopladora 2/imunologia , Brônquios/citologia , Brônquios/imunologia , Células Cultivadas , Humanos , Inflamação/microbiologia , Mitocôndrias/imunologia , Mitocôndrias/microbiologia , Espécies Reativas de Oxigênio/imunologia , Mucosa Respiratória/citologia , Mucosa Respiratória/imunologia , Mucosa Respiratória/microbiologia , Transdução de Sinais
11.
Cytokine ; 110: 306-315, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29807685

RESUMO

Sterile cure from visceralized Leishmania donovani (L. donovani) needs Th1 cell support along with the assistance from innate immune cells, NK cells and NKT cells. NKT cells play as a connecting link between innate and adaptive immune cell and support T helper cell function. Earlier, a categorical function of CD56 positive CD4+ or CD8+ NKT cells was reported in visceral leishmaniasis (VL). It was observed in in vitro that CD4+CD56+NKT cells, but not CD8+CD56+NKT cells, were accumulated at the L. donovani infection site. Therefore, in vitro experiments have been carried out to decipher the mechanism behind preferential accumulation of CD4+CD56+NKT cells at infection site. In this study, 1.89 fold higher expression of CCL4/MIP-1ß was noticed in infected macrophages. The higher expression of CCL4 was correlated with preferential accumulation of CCR5+CD4+CD56+NKT cells and apoptosis of CD8+CD56+NKT cells at in vitro infection site. The CD4+CD56+NKT cells were also observed expressing TGF-ß dominantly. Interaction of CCL4 chemotaxis was interrupted by blocking, which led to drift back the TGF-ß producing CD4+CD56+NKT cells and promoted CD8+CD56+NKT cells recruitment in in vitro infection site. CCR5 blockade also reduced CD25 and FoxP3 positive CD4+CD56+NKT cells in in vitro infection site. Therefore, it was concluded that Leishmania promotes strategic expression of CCL4, which alternately attracts CCR5+ cells, mostly expressing regulatory cytokines, at infection site. This reduces the CD8+CD56+NKT cells at infection site through Smad4 mediated TGF-ß expression and activation of caspases. Data indicates that L. donovani induces higher expression of CCL4 in host cell to attract CCR5+ cells under its strategic plan to downregulate host immune response.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Antígeno CD56/imunologia , Linfócitos T CD8-Positivos/imunologia , Quimiocina CCL4/imunologia , Leishmania donovani/imunologia , Leishmaniose Visceral/imunologia , Células T Matadoras Naturais/imunologia , Adolescente , Adulto , Apoptose/imunologia , Caspases/imunologia , Criança , Feminino , Fatores de Transcrição Forkhead/imunologia , Humanos , Masculino , Pessoa de Meia-Idade , Proteína Smad4/imunologia , Fator de Crescimento Transformador beta/imunologia , Adulto Jovem
12.
Nat Immunol ; 18(9): 995-1003, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28759002

RESUMO

Among the features that distinguish type 1 innate lymphoid cells (ILC1s) from natural killer (NK) cells is a gene signature indicative of 'imprinting' by cytokines of the TGF-ß family. We studied mice in which ILC1s and NK cells lacked SMAD4, a signal transducer that facilitates the canonical signaling pathway common to all cytokines of the TGF-ß family. While SMAD4 deficiency did not affect ILC1 differentiation, NK cells unexpectedly acquired an ILC1-like gene signature and were unable to control tumor metastasis or viral infection. Mechanistically, SMAD4 restrained non-canonical TGF-ß signaling mediated by the cytokine receptor TGFßR1 in NK cells. NK cells from a SMAD4-deficient person affected by polyposis were also hyper-responsive to TGF-ß. These results identify SMAD4 as a previously unknown regulator that restricts non-canonical TGF-ß signaling in NK cells.


Assuntos
Células Matadoras Naturais/citologia , Linfopoese/genética , Proteína Smad4/genética , Fator de Crescimento Transformador beta/imunologia , Polipose Adenomatosa do Colo/genética , Polipose Adenomatosa do Colo/imunologia , Animais , Estudos de Casos e Controles , Diferenciação Celular , Perfilação da Expressão Gênica , Humanos , Imunidade Inata/imunologia , Immunoblotting , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/imunologia , Linfócitos/citologia , Melanoma Experimental/imunologia , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/imunologia , Proteína Smad4/imunologia
13.
Gut ; 66(3): 454-463, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-26681737

RESUMO

OBJECTIVE: Patient-specific (unique) tumour antigens, encoded by somatically mutated cancer genes, generate neoepitopes that are implicated in the induction of tumour-controlling T cell responses. Recent advancements in massive DNA sequencing combined with robust T cell epitope predictions have allowed their systematic identification in several malignancies. DESIGN: We undertook the identification of unique neoepitopes in colorectal cancers (CRCs) by using high-throughput sequencing of cDNAs expressed by standard cancer cell cultures, and by related cancer stem/initiating cells (CSCs) cultures, coupled with a reverse immunology approach not requiring human leukocyte antigen (HLA) allele-specific epitope predictions. RESULTS: Several unique mutated antigens of CRC, shared by standard cancer and related CSC cultures, were identified by this strategy. CD8+ and CD4+ T cells, either autologous to the patient or derived from HLA-matched healthy donors, were readily expanded in vitro by peptides spanning different cancer mutations and specifically recognised differentiated cancer cells and CSC cultures, expressing the mutations. Neoepitope-specific CD8+ T cell frequency was also increased in a patient, compared with healthy donors, supporting the occurrence of clonal expansion in vivo. CONCLUSIONS: These results provide a proof-of-concept approach for the identification of unique neoepitopes that are immunogenic in patients with CRC and can also target T cells against the most aggressive CSC component.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/imunologia , DNA Complementar/análise , Epitopos de Linfócito T/genética , Proteína da Polipose Adenomatosa do Colo/genética , Proteínas de Ciclo Celular/genética , Classe I de Fosfatidilinositol 3-Quinases , Análise Mutacional de DNA , Epitopos de Linfócito T/imunologia , Proteínas F-Box/genética , Proteína 7 com Repetições F-Box-WD , Expressão Gênica , Antígenos HLA/genética , Antígenos HLA/imunologia , Ensaios de Triagem em Larga Escala , Humanos , Células-Tronco Neoplásicas/imunologia , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteína Smad4/genética , Proteína Smad4/imunologia , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Ubiquitina-Proteína Ligases/genética
14.
Cell Death Dis ; 6: e1984, 2015 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-26583325

RESUMO

The transcriptional regulation underlying the differentiation of CD8(+) effector and memory T cells remains elusive. Here, we show that 18-month-old mice lacking the transcription factor Smad4 (homolog 4 of mothers against decapentaplegic, Drosophila), a key intracellular signaling effector for the TGF-ß superfamily, in T cells exhibited lower percentages of CD44(hi)CD8(+) T cells. To explore the role of Smad4 in the activation/memory of CD8(+) T cells, 6- to 8-week-old mice with or without Smad4 in T cells were challenged with Listeria monocytogenes. Smad4 deficiency did not affect antigen-specific CD8(+) T-cell expansion but led to partially impaired cytotoxic function. Less short-lived effector T cells but more memory-precursor effector T cells were generated in the absence of Smad4. Despite that, Smad4 deficiency led to reduced memory CD8(+) T-cell responses. Further exploration revealed that the generation of central memory T cells was impaired in the absence of Smad4 and the cells showed survival issue. In mechanism, Smad4 deficiency led to aberrant transcriptional programs in antigen-specific CD8(+) T cells. These findings demonstrated an essential role of Smad4 in the control of effector and memory CD8(+) T-cell responses to infection.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Proteína Smad4/imunologia , Animais , Diferenciação Celular/imunologia , Memória Imunológica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais , Proteína Smad4/genética
15.
Growth Factors ; 33(3): 169-80, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26066770

RESUMO

Clinical studies showed that renal expression and serum levels of nerve growth factor (NGF) are increased in renal diseases characterized by progressive fibrosis, a pathologic process in which TGF-ß1 mediates most of the key events leading to tubular epithelial-mesenchymal transition (EMT). However, the pathogenic role of high NGF levels has not yet been elucidated. In this study, we found that in tubular renal cells, HK-2, NGF transcriptionally up-regulated TGF-ß1 expression and secretion and enhanced cell motility by activating EMT markers via its receptors, TrkA and p75(NTR). Interestingly, we observed that TGF-ß1-SMAD pathway activation and the up-regulation of EMT markers NGF-induced were both prevented when knockdown of TGF-ß1 gene occurred and that the pretreatment with an antibody anti-NGF reversed the nuclear translocation of pSMAD3/SMAD4 complex. Collectively, our results demonstrated that NGF promotes renal fibrosis via TGF-ß1-signaling activation, suggesting that in kidney diseases high NGF serum levels could contribute to worsen renal fibrosis.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Fibrose/patologia , Túbulos Renais/fisiologia , Fator de Crescimento Neural/metabolismo , Insuficiência Renal Crônica/patologia , Fator de Crescimento Transformador beta1/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Anticorpos/imunologia , Linhagem Celular , Movimento Celular , Células Epiteliais/citologia , Humanos , Fator de Crescimento Neural/sangue , Fator de Crescimento Neural/imunologia , Proteínas do Tecido Nervoso/metabolismo , Interferência de RNA , RNA Interferente Pequeno/genética , Receptor trkA/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Proteína Smad3/antagonistas & inibidores , Proteína Smad3/imunologia , Proteína Smad3/metabolismo , Proteína Smad4/antagonistas & inibidores , Proteína Smad4/imunologia , Proteína Smad4/metabolismo , Transcrição Gênica/genética , Fator de Crescimento Transformador beta1/genética
16.
J Immunol ; 194(5): 2407-14, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25637015

RESUMO

Tissue-resident memory CD8 T cells are a unique subset of virus-specific CTLs that bolster local immune responses after becoming lodged in previously infected tissues. These cells provide enhanced protection by intercepting returning pathogens before a new infection gets established. In contrast, central memory CD8 T cells circulate in the bloodstream and proliferate in secondary lymphoid organs before replenishing effector and memory CD8 T cell populations in remote parts of the body. Both populations of virus-specific memory CD8 T cells participate in immunity to influenza virus infection; however, the signaling pathways that instruct developing memory CD8 T cells to distribute to specific tissues are poorly defined. We show that TGF-ß promotes the development of pulmonary tissue-resident memory T cells via a signaling pathway that does not require the downstream signaling intermediate Sma- and Mad-related protein (Smad)4. In contrast, circulating memory CD8 T cells have no requirement for TGF-ß but show signs of arrested development in the absence of Smad4, including aberrant CD103 expression. These signaling pathways alter the distribution of virus-specific CTLs in the lungs but do not prevent robust cytokine responses. Our data show that Smad4 is required for normal differentiation of multiple subsets of virus-specific CD8 T cells. In normal circumstances, Smad4 may be activated via a pathway that bypasses the TGF-ß receptor. Improved understanding of these signaling pathways could be used to augment vaccine-induced immunity.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linhagem da Célula/imunologia , Memória Imunológica , Pulmão/imunologia , Proteína Smad4/imunologia , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/patologia , Linfócitos T CD8-Positivos/virologia , Diferenciação Celular , Regulação da Expressão Gênica , Vírus da Influenza A/imunologia , Cadeias alfa de Integrinas/genética , Cadeias alfa de Integrinas/imunologia , Pulmão/efeitos dos fármacos , Pulmão/patologia , Pulmão/virologia , Ativação Linfocitária , Camundongos , Camundongos Transgênicos , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/virologia , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/imunologia , Transdução de Sinais , Proteína Smad4/deficiência , Proteína Smad4/genética , Fator de Crescimento Transformador beta/imunologia , Fator de Crescimento Transformador beta/farmacologia , Quimeras de Transplante
17.
Biochem J ; 465(2): 295-303, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25339598

RESUMO

The cAMP signalling pathway plays an essential role in immune functions. In the present study we examined the role of the cAMP/EPAC1 (exchange protein directly activated by cAMP) axis in regulatory T-cell (Treg)-mediated immunosuppression using genetic and pharmacological approaches. Genetic deletion of EPAC1 in Tregs and effector T-cells (Teffs) synergistically attenuated Treg-mediated suppression of Teffs. Mechanistically, EPAC1 inhibition enhanced activation of the transcription factor STAT3 (signal transducer and activator of transcription 3) and up-regulated SMAD7 expression while down-regulating expression of SMAD4. Consequently, CD4+ T-cells were desensitized to transforming growth factor (TGF) ß1, a cytokine employed by Tregs to exert a broad inhibitory function within the immune system. Furthermore, deletion of EPAC1 led to production of significant levels of ovalbumin IgG antibodies in a low-dose, oral-tolerance mouse model. These in vivo observations are consistent with the finding that EPAC1 plays an important role in Treg-mediated suppression. More importantly, pharmacological inhibition of EPAC1 using an EPAC-specific inhibitor recapitulates the EPAC1 deletion phenotype both in vivo and in vitro. The results of the present study show that EPAC1 boosts Treg-mediated suppression, and identifies EPAC1 as a target with broad therapeutic potential because Tregs are involved in numerous pathologies, including autoimmunity, infections and a wide range of cancers.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/imunologia , Tolerância Imunológica/fisiologia , Linfócitos T Reguladores/imunologia , Animais , Fatores de Troca do Nucleotídeo Guanina/genética , Camundongos , Camundongos Knockout , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/imunologia , Proteína Smad4/genética , Proteína Smad4/imunologia , Proteína Smad7/genética , Proteína Smad7/imunologia , Linfócitos T Reguladores/citologia , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/imunologia
18.
Parasitol Res ; 113(10): 3745-57, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25039015

RESUMO

Smad family proteins are essential cellular mediators of the transforming growth factor-ß superfamily. In the present study, we identified two members of the Smad proteins, Smad8 and Smad4 homologues (termed as EgSmadE and EgSmadD, respectively), from Echinococcus granulosus, the causative agent of cystic echinococcosis (CE). Phylogenetic analysis placed EgSmadE in the Smad1, 5, and 8 subgroup of the R-Smad sub-family and EgSmadD in the Co-Smad family. Furthermore, EgSmadE and EgSmadD attained a high homology to EmSmadE and EmSmadD of E. multilocularis, respectively. Both EgSmadE and EgSmadD were co-expressed in the larval stages and exhibited the highest transcript levels in activated protoscoleces, and their encoded proteins were co-localized in the sub-tegumental and tegumental layer of the parasite. As shown by yeast two-hybrid and pull-down analysis, EgSmadE displayed a positive binding interaction with EgSmadD. In addition, EgSmadE localized in the nuclei of Mv1Lu cells (mink lung epithelial cells) upon treatment with human TGF-ß1 or human BMP2, indicating that EgSmadE is capable of being translocated into nucleus, in vitro. Our study suggests that EgSmadE and EgSmadD may take part in critical biological processes, including echinococcal growth, development, and parasite-host interaction.


Assuntos
Equinococose/parasitologia , Echinococcus granulosus/genética , Transdução de Sinais , Proteína Smad4/genética , Proteína Smad8/genética , Animais , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/metabolismo , Linhagem Celular , DNA de Helmintos/química , DNA de Helmintos/genética , Echinococcus granulosus/classificação , Echinococcus granulosus/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Genoma Helmíntico/genética , Proteínas de Helminto/genética , Proteínas de Helminto/imunologia , Proteínas de Helminto/metabolismo , Interações Hospedeiro-Parasita , Humanos , Soros Imunes/imunologia , Filogenia , Coelhos , Proteína Smad4/imunologia , Proteína Smad4/metabolismo , Proteína Smad8/imunologia , Proteína Smad8/metabolismo , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo
19.
Arch Oral Biol ; 58(11): 1744-50, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23827715

RESUMO

INTRODUCTION: During root formation, Smad-4 plays a key role during the epithelial-mesenchymal interactions and the Hertwig's epithelial root sheath (HERS) apical proliferation. The root formation and eruption of rat molars is impeded by alendronate treatment due to the inhibition of bone resortion by this drug. The present study aimed to examine the structures affected in the developing root and immunodetect the presence of Smad-4 in rats treated with alendronate. METHODS: Newborn Wistar rats were daily injected 2.5 mg/kg alendronate (ALN) during 9, 12 and 30 days. The controls (CON) were injected with saline. The maxillae were fixed and embedded in paraffin or Spurr resin. Paraffin sections were incubated in Smad-4 antibody that was labelled with DAB. The ultrathin sections were examined in a transmission electron microscope. RESULTS: In ALN, a short portion of root dentine was formed; the epithelial diaphragm (ED) and the dental follicle (DF) were disorganized by the contact of bone trabeculae. The (CON) molar roots developed normally. Smad-4 labelling was detected in the cytoplasm of fibroblasts and cementoblasts adjacent to the cementum in CON; in ALN group, few ED cells presented weak immunolabelling. Ultrastructurally, the ED and DF appeared disrupted due to the presence of thin bone trabeculae between its cells. It resulted in the lack of apical proliferation of HERS and, consequently, arrest of root formation. CONCLUSION: The immunodetection of Smad-4 in the DF cells of ALN specimens indicates that the signalling for the differentiation of these cells into cementum-forming fibroblasts and cementoblasts occurs, despite the impairment of root elongation.


Assuntos
Alendronato/efeitos adversos , Reabsorção Óssea/etiologia , Cementogênese/efeitos dos fármacos , Cemento Dentário/efeitos dos fármacos , Dente Molar/efeitos dos fármacos , Proteína Smad4/análise , Raiz Dentária/efeitos dos fármacos , Animais , Proteínas Morfogenéticas Ósseas/efeitos dos fármacos , Proteínas Morfogenéticas Ósseas/metabolismo , Diferenciação Celular/efeitos dos fármacos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Microscopia Eletrônica , Dente Molar/citologia , Dente Molar/crescimento & desenvolvimento , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Proteína Smad4/imunologia , Raiz Dentária/citologia , Raiz Dentária/crescimento & desenvolvimento , Fator de Crescimento Transformador beta1/efeitos dos fármacos , Fator de Crescimento Transformador beta1/metabolismo
20.
Cell Signal ; 24(10): 1923-30, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22735812

RESUMO

Activation of hepatic stellate cells (HSC) plays a pivotal role in the development of hepatic fibrosis. Transforming growth factor-ß1 (TGF-ß1) is considered to be the main stimuli factor responsible for the activation of HSC. MicroRNAs (miRNAs) have recently been shown to regulate cell proliferation, differentiation, and apoptosis. The involvement of miRNAs and their roles in TGF-ß1-induced HSC activation remains largely unknown. Our study found that the expression of miR-146a was downregulated in HSC in response to TGF-ß1 stimulation in dose-dependent manner by one-step real-time quantitative PCR. Moreover, we sought to examine whether miR-146a became dysregulated in CCl(4)-induced hepatic fibrosis in rats. Our study revealed that miR-146a was downregulated in liver fibrotic tissues. In addition, The HSC transfected with miR-146a mimics exhibited attendated TGF-ß1-induced α-smooth muscle actin (α-SMA) expression compared with the control. Furthermore, overexpression of miR-146a suppressed TGF-ß-induced HSC proliferation, and increased HSC apoptosis. Bioinformatics analyses predict that SMAD4 is the potential target of miR-146a. MiR-146a overexpression in TGF-ß1-treated HSC did not decrease target mRNA levels, but significantly reduced target protein expression. These results suggested that miR-146a may function as a novel regulator to modulate HSC activation during TGF-ß1 induction by targeting SMAD4.


Assuntos
Proliferação de Células , Regulação da Expressão Gênica , Células Estreladas do Fígado/citologia , MicroRNAs/imunologia , Proteína Smad4/genética , Fator de Crescimento Transformador beta1/imunologia , Actinas/genética , Animais , Apoptose , Linhagem Celular , Células Estreladas do Fígado/imunologia , Células Estreladas do Fígado/metabolismo , Células Estreladas do Fígado/patologia , Fígado/imunologia , Fígado/metabolismo , Fígado/patologia , Cirrose Hepática/genética , Cirrose Hepática/imunologia , Cirrose Hepática/patologia , Masculino , MicroRNAs/genética , Ratos , Ratos Sprague-Dawley , Proteína Smad4/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...