Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(18)2023 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-37761990

RESUMO

Recent studies have highlighted the therapeutic potential of small extracellular bodies derived from mesenchymal stem cells (MSC-sEVs) for various diseases, notably through their ability to alter T-cell differentiation and function. The current study aimed to explore immunomodulatory pathway alterations within T cells through mRNA sequencing of activated T cells cocultured with bone marrow-derived MSC-sEVs. mRNA profiling of activated human T cells cocultured with MSC-sEVs or vehicle control was performed using the QIAGEN Illumina sequencing platform. Pathway networks and biological functions of the differentially expressed genes were analyzed using Ingenuity pathway analysis (IPA)® software, KEGG pathway, GSEA and STRING database. A total of 364 differentially expressed genes were identified in sEV-treated T cells. Canonical pathway analysis highlighted the RhoA signaling pathway. Cellular development, movement, growth and proliferation, cell-to-cell interaction and inflammatory response-related gene expression were altered. KEGG enrichment pathway analysis underscored the apoptosis pathway. GSEA identified enrichment in downregulated genes associated with TNF alpha and interferon gamma response, and upregulated genes related to apoptosis and migration of lymphocytes and T-cell differentiation gene sets. Our findings provide valuable insights into the mechanisms by which MSC-sEVs implement immunomodulatory effects on activated T cells. These findings may contribute to the development of MSC-sEV-based therapies.


Assuntos
Vesículas Extracelulares , Fator de Necrose Tumoral alfa , Humanos , Fator de Necrose Tumoral alfa/genética , Interferon gama , Linfócitos T , Apoptose/genética
2.
Curr Opin Clin Nutr Metab Care ; 25(3): 209-215, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-35762173

RESUMO

PURPOSE OF REVIEW: Milk-derived extracellular vesicles (MDEVs) are nanovesicles that carry microRNA (miRNA) DNA, RNA, proteins and lipids. MDEVs have a potential of therapeutic targets, based on their properties and cargo profile. The present review summarizes recent studies on MDEVs, their cargo and potential role in mammalian development. RECENT FINDINGS: The detailed characterization of their miRNA cargo leads to the conclusion of their potential importance in the regulation of gene expression, immune function, development and infant growth.While their miRNAs are important regulatory elements and their profile expression was characterized in various mammalian milk sources, little is known about their effect on infant health and development. MiRNA activity in breast milk is likely influenced by the overall ecosystem of the early environment, including maternal characteristics, behaviors, and health. SUMMARY: MDEVs may have an important role in early child development and infant future health. Understanding benefits of MDEVs characteristics have potential role on gut maturation, immune system development and the prevention of metabolic disorders.


Assuntos
Vesículas Extracelulares , MicroRNAs , Leite Humano , Meio Ambiente , Vesículas Extracelulares/metabolismo , Feminino , Humanos , Lactente , MicroRNAs/metabolismo , Leite Humano/metabolismo
3.
J Pediatr Gastroenterol Nutr ; 74(1): e8-e15, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34371509

RESUMO

OBJECTIVE: The highly expressed microRNAs (miRNAs) in milk are known as beneficial miRNAs, such as mir148a-3p, which is related to immune system development and disease prevention. There is a need to study their expression and secretion regulatory mechanism in breast milk. We hypothesize that oxytocin can be involved in the regulation of expression and secretion of milk-derived miRNAs. METHODS: Initially, oxytocin's effect on miRNA expression in human mammary cells was analyzed. Secondly, the expression of selected miRNAs in mothers' colostrum treated or not with oxytocin before, during, or after labor was compared. MiRNA expression was analyzed by quantitative real-time PCR. RESULTS: The expression of miR-148a was significantly upregulated, and miR-320 downregulated in oxytocin-treated mammary cells as well as their secreted extracellular vesicles to the media, compared with untreated cells. MiR-148a was found to be upregulated, and miR-320 was downregulated in the human colostrum of exogenous oxytocin-treated mothers. Moreover, miR-320 was highly expressed compared with miR-148a in the colostrum of mothers that did not receive exogenous oxytocin. In contrast, in the milk of mothers who received exogenous oxytocin, the expression of miRNA-148-3p was highly expressed compared with miR-320. CONCLUSIONS: This study shows that oxytocin modulates the expression of main milk-derived miRNAs. Our findings provide a novel insight into oxytocin's role in milk composition by regulating miRNA expression. Our results implicate that oxytocin increases miRNA expression in mammary epithelial cells and human milk, affecting human milk composition and may contribute to further infant health.


Assuntos
Vesículas Extracelulares , MicroRNAs , Colostro/química , Vesículas Extracelulares/química , Feminino , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Leite Humano/química , Ocitocina/metabolismo , Gravidez
4.
Nutrients ; 12(9)2020 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-32858892

RESUMO

The aim of this study was to investigate the therapeutic effect of cow and human milk derived exosomes (MDEs) on colitis. We used gavage administration of fluorescent labeled MDEs to track their localization patterns in vivo and studied their therapeutic effect on colitis in a dextran sulfate sodium (DSS)-induced colitis model. MDEs attenuated the severity of colitis induced by DSS and statistically reduced the histopathological scoring grade and shortening of the colon. Likewise, treatment with MDEs reduced the expression of interleukin 6 and tumor necrosis factor-α. Moreover, miRNAs highly expressed in milk, such as miRNA-320, 375, and Let-7, were found to be more abundant in the colon of MDE-treated mice compared with untreated mice; contrastingly, the expression of their target genes, mainly DNA methyltransferase 1 (DNMT1) and DNMT3 were downregulated. Furthermore, the level of TGF-ß was upregulated in the colon of MDE-treated mice. We demonstrated that MDEs have a therapeutic and anti-inflammatory effect on colitis, involving several complementary pathways in its mechanism of action. The therapeutic effects of MDEs might have implications for the possible addition of MDEs as a nutrient in enteral nutrition formulas for patients with inflammatory bowel disease.


Assuntos
Colite/terapia , Exossomos/metabolismo , Leite Humano/metabolismo , Leite/metabolismo , Animais , Sulfato de Dextrana , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos BALB C
5.
J Transl Med ; 17(1): 325, 2019 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-31564251

RESUMO

BACKGROUND: Breastfeeding is the ideal source of infant nutrition. Human milk consists not only of nutrients but also biologically active components. Among these latter compounds, exosomes contain proteins, lipids, mRNAs and miRNAs. METHODS: To elucidate the biological effects of milk-derived exosomes (MDEs) on normal colonic epithelial cells compared to colonic tumor cells, we incubated cells with MDEs. MDEs were able to enter into normal and tumor cells and change their miRNA expression profiles. Proliferation, cell morphology and protein expression were analyzed in these cells. RESULTS: Human milk-derived exosomes induced proliferation- and epithelial mesenchymal transformation-related changes, such as collagen type I and twist expression, in normal but not in tumor cells. PTEN, a target of miRNA-148a, was downregulated in normal but not in tumor cells following incubation with MDEs. Moreover, miRNA-148a-3p knockdown cells were used to demonstrate the importance of miRNA in the effect of exosomes on cell proliferation and protein expression. MDEs inhibited proliferation and DNMT1 expression in cells with knockdown of miRNA-148a. CONCLUSIONS: In conclusion, the positive effect of exosomes on normal cells without affecting tumor cells may presents an aspect of their safety when considering it use as a nutritional supplement to infant formula.


Assuntos
Colo/citologia , Neoplasias do Colo/patologia , Células Epiteliais/citologia , Exossomos/metabolismo , Feto/citologia , MicroRNAs/metabolismo , Leite Humano/metabolismo , Animais , Bovinos , Linhagem Celular Tumoral , Proliferação de Células , Forma Celular , DNA (Citosina-5-)-Metiltransferase 1/genética , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , Regulação para Baixo/genética , Humanos , MicroRNAs/genética , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
6.
Mol Nutr Food Res ; 61(10)2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28643865

RESUMO

SCOPE: Breastfeeding is associated with reduced risk of infection, immune-mediated disorders, obesity, and even cancer. Recently it was found that breast milk contains a variety of microRNAs (miRNAs) in the skim and fat layer that can be transferred to infants, and appear to play important roles in those biological functions. METHODS AND RESULTS: This study applied next generation sequencing and quantitative real-time PCR analysis to determine the miRNA expression profile of the skim and fat fraction of human, goat, and bovine milk as well as infant formulas. Human and mammalian milk were found to contain known advantageous miRNAs in exosomes and also in the fat layer. These miRNAs are highly conserved in human, bovine and goat milk. However, they were not detected in several infant formulas. Further, miRNAs present in milk were able to enter normal and tumor cells and affect their biological functions. Following incubation of milk derived human miRNA with normal and cancer cells, the expression of miRNA-148a was upregulated and the expression of the DNA methyltransferase1 target gene of miRNA-148a was down regulated. CONCLUSION: These results reinforce previous findings on the importance of miRNA in breast milk. Future studies should concentrate on the addition of miRNA to infant formulas.


Assuntos
MicroRNAs/isolamento & purificação , Leite Humano/química , Leite/química , Animais , Bovinos , Linhagem Celular Tumoral , Células Cultivadas , Biologia Computacional , Células Epiteliais/metabolismo , Exossomos/genética , Exossomos/metabolismo , Cabras , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Fórmulas Infantis/química , Pasteurização , Análise de Sequência de RNA , Transcriptoma
7.
PLoS One ; 10(5): e0126730, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25951185

RESUMO

High doses of bleomycin administered to patients with lymphomas and other tumors lead to significant lung toxicity in general, and to apoptosis of epithelial cells, in particular. Apoptosis of alveolar epithelium is an important step in the pathogenesis of bleomycin-induced pulmonary fibrosis. The Fas-FasL pathway is one of the main apoptotic pathways involved. Telomerase is a ribonucleoprotein RNA-dependent DNA polymerase complex consisting of an RNA template and a catalytic protein, telomerase reverse transcriptase (TERT). Telomerase also possess extra-telomeric roles, including modulation of transcription of anti-apoptotic genes, differentiation signals, and more. We hypothesized that telomerase overexpression affects Fas-induced epithelial cell apoptosis by an extra-telomeric role such as regulation of anti-apoptotic genes, specifically FLICE-like inhibitory protein (FLIP). Telomerase in mouse (MLE) and human (A549) lung epithelial cell lines was upregulated by transient transfection using cDNA hTERT expression vector. Telomerase activity was detected using a real-time PCR-based system. Bleomycin, and bleomycin-induced Fas-mediated apoptosis following treatment with anti-Fas activating mAb or control IgG, were assessed by Annexin V staining, FACS analysis, and confocal microscopy; caspase cleavage by Western blot; FLIP or Fas molecule detection by Western blot and flow cytometry. hTERT transfection of lung epithelial cells resulted in a 100% increase in their telomerase activity. Fas-induced lung epithelial cell apoptosis was significantly reduced in hTERT-transfected cells compared to controls in all experiments. Lung epithelial cells with increased telomerase activity had higher levels of FLIP expression but membrane Fas expression was unchanged. Upregulation of hTERT+ in human lung epithelial cells and subsequent downregulation of FLIP by shFLIP-RNA annulled hTERT-mediated resistance to apoptosis. Telomerase-mediated FLIP overexpression may be a novel mechanism to confer protection from apoptosis in bleomycin-exposed human lung epithelial cells.


Assuntos
Antibióticos Antineoplásicos/efeitos adversos , Bleomicina/efeitos adversos , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Células Epiteliais/efeitos dos fármacos , Telomerase/genética , Receptor fas/metabolismo , Animais , Apoptose/efeitos dos fármacos , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Linhagem Celular , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Humanos , Pulmão/citologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Telomerase/metabolismo , Transfecção , Regulação para Cima
8.
Nucleic Acids Res ; 43(10): 5130-44, 2015 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-25908786

RESUMO

Cancer is a complex disease that involves aberrant gene expression regulation. Discriminating the modified expression patterns driving tumor biology from the many that have no or little contribution is important for understanding cancer molecular basis. Recurrent deregulation patterns observed in multiple cancer types are enriched for such driver events. Here, we studied splicing alterations in hundreds of matched tumor and normal RNA-seq samples of eight solid cancer types. We found hundreds of cassette exons for which splicing was altered in multiple cancer types and identified a set of highly frequent altered splicing events. Specific splicing regulators, including RBFOX2, MBNL1/2 and QKI, appear to account for many splicing alteration events in multiple cancer types. Together, our results provide a first global analysis of regulated splicing alterations in cancer and identify common events with a potential causative role in solid tumor development.


Assuntos
Processamento Alternativo , Neoplasias/genética , Éxons , Humanos , Neoplasias/metabolismo , Fatores de Processamento de RNA , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Sequências Reguladoras de Ácido Ribonucleico , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo
9.
J Autoimmun ; 59: 67-76, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25812467

RESUMO

Immune cells, particularly those expressing the ligand of the Fas-death receptor (FasL), e.g. cytotoxic T cells, induce apoptosis in 'undesirable' self- and non-self-cells, including lung fibroblasts, thus providing a means of immune surveillance. We aimed to validate this mechanism in resolution of lung fibrosis. In particular, we elucidated whether FasL(+) immune cells possess antifibrotic capabilities by induction of FasL-dependent myofibroblast apoptosis and whether antagonists of membrane (m) and soluble (s) FasL can inhibit these capabilities. Myofibroblast interaction with immune cells and its FasL-dependency, were investigated in vitro in coculture with T cells and in vivo, following transplantation into lungs of immune-deficient syngeneic Rag-/- as well as allogeneic SCID mice, and into lungs and air pouches of FasL-deficient (gld) mice, before and after reconstitution of the mice with wild-type (wt), FasL(+) immune cells. We found that myofibroblasts from lungs resolving fibrosis undergo FasL-dependent T cell-induced apoptosis in vitro and demonstrate susceptibility to in vivo immune surveillance in lungs of reconstituted, immune- and FasL-deficient, mice. However, immune-deficient Rag-/- and SCID mice, and gld-mice with FasL-deficiency, endure the accumulation of transplanted myofibroblasts in their lungs with subsequent development of fibrosis. Concomitantly, gld mice, in contrast to chimeric FasL-deficient mice with wt immune cells, accumulated transplanted myofibroblasts in the air pouch model. In humans we found that myofibroblasts from fibrotic lungs secrete sFasL and resist T cell-induced apoptosis, whereas normal lung myofibroblasts are susceptible to apoptosis but acquire resistance upon addition of anti-s/mFasL to the coculture. Immune surveillance, particularly functional FasL(+) immune cells, may represent an important extrinsic component in myofibroblast apoptosis and serve as a barrier to fibrosis. Factors interfering with Fas/FasL-immune cell-myofibroblast interaction such as sFasL secreted by fibrotic-lung myofibroblasts, may abrogate immune surveillance during fibrosis. Annulling these factors may pave a new direction to control human lung fibrosis.


Assuntos
Apoptose , Proteína Ligante Fas/metabolismo , Pulmão/patologia , Miofibroblastos/metabolismo , Linfócitos T Citotóxicos/imunologia , Transferência Adotiva , Animais , Anticorpos Bloqueadores/farmacologia , Apoptose/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Proteína Ligante Fas/genética , Fibrose , Genes RAG-1/genética , Humanos , Vigilância Imunológica/efeitos dos fármacos , Pulmão/imunologia , Camundongos , Camundongos Knockout , Camundongos SCID , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/patologia , Linfócitos T Citotóxicos/transplante
10.
Cell Rep ; 7(2): 501-513, 2014 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-24726367

RESUMO

The kinase Mnk2 is a substrate of the MAPK pathway and phosphorylates the translation initiation factor eIF4E. In humans, MKNK2, the gene encoding for Mnk2, is alternatively spliced yielding two splicing isoforms with differing last exons: Mnk2a, which contains a MAPK-binding domain, and Mnk2b, which lacks it. We found that the Mnk2a isoform is downregulated in breast, lung, and colon tumors and is tumor suppressive. Mnk2a directly interacts with, phosphorylates, activates, and translocates p38α-MAPK into the nucleus, leading to activation of its target genes, increasing cell death and suppression of Ras-induced transformation. Alternatively, Mnk2b is pro-oncogenic and does not activate p38-MAPK, while still enhancing eIF4E phosphorylation. We further show that Mnk2a colocalization with p38α-MAPK in the nucleus is both required and sufficient for its tumor-suppressive activity. Thus, Mnk2a downregulation by alternative splicing is a tumor suppressor mechanism that is lost in some breast, lung, and colon tumors.


Assuntos
Processamento Alternativo , Núcleo Celular/metabolismo , Transformação Celular Neoplásica/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Camundongos , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Proteínas ras/metabolismo
11.
Clin Exp Metastasis ; 30(1): 103-17, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22806361

RESUMO

We previously found LOXL4 to be alternatively spliced in an anatomic site-specific manner in tumors involving the serosal cavities. LOXL4 splice variants were predominantly or exclusively expressed in effusion specimens from ovarian and breast carcinoma patients, and were absent in primary carcinomas. In the present study, LOXL4 full-length or splice variants were overexpressed in ES-2 and MDA-MB-231 cells and their invasive and metastatic potential and microRNA expression profile were evaluated. ES-2 cells were further injected into SCID mice ovaries and the extent of tumor progression and metastases formation were compared. We show that both splice variants have a positive effect on the metastatic potential of cells in vitro and on tumor progression in vivo. In contrast, full-length LOXL4 is not pro-metastatic, and may even be considered as a tumor suppressor. In addition, we show that LOXL4 is a possible splicing target of the oncogenic splicing factors SRSF1 and hnRNP A1. In conclusion, our results point to a significant role for LOXL4 alternative splicing in tumor progression.


Assuntos
Processamento Alternativo , Aminoácido Oxirredutases/genética , Neoplasias da Mama/genética , Neoplasias Ovarianas/genética , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Western Blotting , Neoplasias da Mama/secundário , Feminino , Perfilação da Expressão Gênica , Ribonucleoproteína Nuclear Heterogênea A1 , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/genética , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/metabolismo , Humanos , Técnicas Imunoenzimáticas , Imunoprecipitação , Isoenzimas , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos SCID , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias Ovarianas/secundário , Mutação Puntual/genética , Complexo de Endopeptidases do Proteassoma/química , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteína-Lisina 6-Oxidase , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Processamento de Serina-Arginina , Esferoides Celulares , Células Tumorais Cultivadas
12.
J Pathol ; 229(4): 630-9, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23132731

RESUMO

An increasing body of evidence connects alterations in the process of alternative splicing with cancer development and progression. However, a direct role of splicing factors as drivers of cancer development is mostly unknown. We analysed the gene copy number of several splicing factors in colon and lung tumours, and found that the gene encoding for the splicing factor SRSF6 is amplified and over-expressed in these cancers. Moreover, over-expression of SRSF6 in immortal lung epithelial cells enhanced proliferation, protected them from chemotherapy-induced cell death and converted them to be tumourigenic in mice. In contrast, knock-down of SRSF6 in lung and colon cancer cell lines inhibited their tumourigenic abilities. SRSF6 up- or down-regulation altered the splicing of several tumour suppressors and oncogenes to generate the oncogenic isoforms and reduce the tumour-suppressive isoforms. Our data suggest that the splicing factor SRSF6 is an oncoprotein that regulates the proliferation and survival of lung and colon cancer cells.


Assuntos
Adenocarcinoma/genética , Neoplasias da Mama/genética , Neoplasias do Colo/genética , Neoplasias Pulmonares/genética , Proteínas Nucleares/genética , Proteínas Oncogênicas/genética , Fosfoproteínas/genética , Proteínas de Ligação a RNA/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Processamento Alternativo , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Carcinoma Ductal/genética , Carcinoma Ductal/metabolismo , Carcinoma Ductal/patologia , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Transformação Celular Neoplásica , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Progressão da Doença , Regulação para Baixo , Feminino , Dosagem de Genes , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas/metabolismo , Fosfoproteínas/metabolismo , Isoformas de Proteínas , Splicing de RNA , Proteínas de Ligação a RNA/metabolismo , Fatores de Processamento de Serina-Arginina , Regulação para Cima
13.
Am J Respir Cell Mol Biol ; 47(3): 271-9, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22582174

RESUMO

A prominent feature of fibrotic tissue in general and of lungs in particular is fibroblast proliferation and accumulation. In patients overcoming fibrosis, apoptosis limits this excessive cell growth. We have previously shown resistance to Fas-induced apoptosis of primary lung fibroblasts from mice with bleomycin-induced lung fibrosis, their escape from immune surveillance, and continued accumulation in spite of overexpression of the Fas death receptor. Cellular FLICE-like inhibitory protein (c-FLIP) is a regulator of cell death receptor-induced apoptosis in many cell types. We aimed to determine c-FLIP levels in myofibroblasts from fibrotic lungs and to directly assess c-FLIP's role in apoptosis and proliferation of primary lung myofibroblasts. c-FLIP levels were determined by apoptosis gene array, flow cytometry, Western blot, and immunofluorescence before and after down-regulation with a specific small interfering RNA. Apoptosis was assessed by caspase cleavage in Western blot and by Annexin V affinity labeling after FACS and tissue immunofluorescence. Proliferation was assessed by BrdU uptake, also using FACS and immunofluorescence. We show that myofibroblasts from lungs of humans with idiopathic pulmonary fibrosis and from bleomycin-treated versus normal saline-treated mice up-regulate c-FLIP levels. Using the animal model, we show that fibrotic lung myofibroblasts divert Fas signaling from apoptosis to proliferation and that this requires signaling by TNF receptor-associated factor (TRAF) and NF-κB. c-FLIP down-regulation reverses the effect of Fas activation, causing increased apoptosis, decreased proliferation, and diminished recruitment of TRAF to the DISC complex. This indicates that c-FLIP is essential for myofibroblast accumulation and may serve as a potential target to manipulate tissue fibrosis.


Assuntos
Apoptose/fisiologia , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/fisiologia , Proliferação de Células , Miofibroblastos/patologia , Fibrose Pulmonar/patologia , Receptor fas/fisiologia , Animais , Anexina A5/metabolismo , Sequência de Bases , Western Blotting , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Caspases/metabolismo , Primers do DNA , Citometria de Fluxo , Imunofluorescência , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais
14.
Cancer Res ; 71(13): 4464-72, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21586613

RESUMO

The process of alternative splicing is widely misregulated in cancer, but the contribution of splicing regulators to cancer development is largely unknown. In this study, we found that the splicing factor hnRNP A2/B1 is overexpressed in glioblastomas and is correlated with poor prognosis. Conversely, patients who harbor deletions of the HNRNPA2B1 gene show better prognosis than average. Knockdown of hnRNP A2/B1 in glioblastoma cells inhibited tumor formation in mice. In contrast, overexpression of hnRNP A2/B1 in immortal cells led to malignant transformation, suggesting that HNRNPA2B1 is a putative proto-oncogene. We then identified several tumor suppressors and oncogenes that are regulated by HNRNPA2B1, among them are c-FLIP, BIN1, and WWOX, and the proto-oncogene RON. Knockdown of RON inhibited hnRNP A2/B1 mediated transformation, which implied that RON is one of the mediators of HNRNPA2B1 oncogenic activity. Together, our results indicate that HNRNPA2B1 is a novel oncogene in glioblastoma and a potential new target for glioblastoma therapy.


Assuntos
Neoplasias Encefálicas/genética , Genes Supressores de Tumor , Glioblastoma/genética , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/genética , Processamento Alternativo , Animais , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Dosagem de Genes , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Glioblastoma/metabolismo , Glioblastoma/patologia , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/biossíntese , Humanos , Camundongos , Células NIH 3T3 , Proto-Oncogene Mas , Receptores Proteína Tirosina Quinases/genética , Regulação para Cima
15.
Proc Natl Acad Sci U S A ; 104(51): 20460-5, 2007 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-18077384

RESUMO

Tissue fibrosis evolving from impaired tissue remodeling after injury is characterized by myofibroblast accumulation. We propose that during the development of fibrosis myofibroblasts acquire an immune-privileged cell phenotype, allowing their uninterrupted accumulation. Using the murine model of bleomycin-induced lung fibrosis in mice, we show that myofibroblasts that accumulate in lungs with fibrosis, but not in normal lungs, kill Fas(+) lymphocytes, resist Fas-induced apoptosis, and survive longer when grafted into allogeneic mice. In contrast, bleomycin-treated FasLigand (FasL)-deficient (gld) chimeric mice did not accumulate myofibroblasts or collagen in their lungs, and their FasL(-) myofibroblasts did not survive after alloengraftment. This finding indicates that myofibroblasts possess Fas/FasL-pathway-dependent characteristics that allow them to escape from immune surveillance and resulting organ fibrosis.


Assuntos
Fibroblastos/imunologia , Vigilância Imunológica , Pneumopatias/imunologia , Pneumopatias/patologia , Mioblastos/imunologia , Actinas/metabolismo , Animais , Apoptose , Colágeno/metabolismo , Proteína Ligante Fas/genética , Fibrose , Pneumopatias/induzido quimicamente , Linfócitos/imunologia , Camundongos , Camundongos Mutantes , Receptor fas/genética , Receptor fas/metabolismo
16.
Am J Respir Cell Mol Biol ; 36(3): 270-5, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16990614

RESUMO

The Fas/Fas ligand (FasL) apoptotic pathway has been shown to be involved in bleomycin-induced lung fibrosis. We examined the hypothesis that myofibroblasts from fibrotic lungs possess a cytotoxic phenotype that causes apoptosis of epithelial cells via the Fas/FasL pathway. We show in vivo epithelial cell apoptosis and associated upregulation of Fas and apoptotic Fas pathway genes in epithelial cells of lungs with bleomycin-induced fibrosis. In addition, we show that FasL surface molecules are overexpressed on alpha-SMA-positive cells in mice with bleomycin-induced fibrosis, and in humans with idiopathic pulmonary fibrosis. This enables the molecules to kill Fas-positive epithelial cells. In contrast, FasL-deficient myofibroblasts lose this myofibroblast cytotoxic phenotype, both in vivo and in vitro. In vivo, there was no bleomycin-induced epithelial cell apoptosis, as assessed by specific M30 staining in chimeric FasL-deficient mice that lacked FasL-positive myofibroblasts. In vitro, FasL-positive, but not FasL-negative myofibroblasts, induce mouse lung epithelial cell apoptosis. Thus myofibroblast cytotoxicity may underlie the absence of re-epithelialization, resulting in persistent lung fibrosis.


Assuntos
Apoptose , Células Epiteliais/citologia , Proteína Ligante Fas/metabolismo , Fibroblastos/patologia , Fibrose Pulmonar/patologia , Animais , Apoptose/efeitos dos fármacos , Bleomicina/farmacologia , Quimera , Técnicas de Cocultura , Células Epiteliais/efeitos dos fármacos , Proteína Ligante Fas/deficiência , Proteína Ligante Fas/genética , Fibroblastos/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Humanos , Linfócitos/citologia , Linfócitos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptor fas/genética , Receptor fas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...