Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
J Physiol ; 597(12): 3107-3131, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31026345

RESUMO

KEY POINTS: In muscular cells, eukaryotic initiation factor subunit f (eIF3f) activates protein synthesis by allowing physical interaction between mechanistic target of rapamycin complex 1 (MTORC1) and ribosomal protein S6 kinase 1 (S6K1), although its physiological role in animals is unknown. A knockout approach suggests that homozygous mice carrying a null mutation of the eIF3f gene fail to develop and consequently die at early embryonic stage, whereas heterozygous mice associated with a partial depletion of eIF3f gene grow normally and are phenotypically indistinguishable from wild-type mice. Heterozygous mice express reduced eIF3f mRNA and protein levels in skeletal muscles and show diminished muscle mass associated with a decrease in the protein synthesis rate and an inhibition of the MTORC1 pathway. During hindlimb immobilization, heterozygous eIF3f mice display an exacerbated immobilization-induced muscle atrophy associated with reduced protein synthesis. These results highlight the essential role of eIF3f during embryonic development and its involvement in muscular homeostasis via protein synthesis regulation. ABSTRACT: Eukaryotic translation initiation factor 3, subunit F (eIF3f), a component of eIF3 complex, plays an important role in protein synthesis regulation, although its physiological functions are unknown. We generated and analysed mice carrying a null mutation in the eIF3f gene. We showed that homozygous eIF3f knockout fail to develop and that eIF3f-/- embryos die at an early stage of development but after the pre-implantation stage. However, disrupting one eIF3f allele does not affect growth, viability and fertility of heterozygous mice but, instead, reduces eIF3f mRNA and protein levels in all tissues examined. Although heterozygous mice are phenotypically indistinguishable from wild-type mice, they present a diminished body weight and a lean mass reduction associated with normal body size. Interestingly, skeletal muscles are mainly affected and display an altered cell size without modification of fibre number. Skeletal muscles of heterozygous mice show a deficiency in polysome content, a decrease in protein synthesis rate and an inhibition of the mechanistic target of rapamycin (MTOR) pathway. We then studied the effects of hindlimb immobilization that mimic muscle disuse on heterozygous mice aiming to further explore the involvement of eIF3f in protein synthesis. We found that eIF3f partial depletion amplifies muscle atrophy compared to wild-type mice. Mass and cross-sectional area decreases were associated with reduced MTOR pathway activation and protein synthesis rate. Taken together, our data indicate that eIF3f is essential for mice embryonic development and controls adult skeletal muscle mass via protein synthesis regulation in a MTOR-dependent manner.


Assuntos
Desenvolvimento Embrionário , Fator de Iniciação 3 em Eucariotos/genética , Músculo Esquelético/patologia , Atrofia Muscular/genética , Animais , Fator de Iniciação 3 em Eucariotos/metabolismo , Feminino , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Músculo Esquelético/metabolismo , Atrofia Muscular/metabolismo
2.
BMC Biol ; 16(1): 65, 2018 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-29895328

RESUMO

BACKGROUND: Skeletal muscle atrophy is a common feature of numerous chronic pathologies and is correlated with patient mortality. The REDD1 protein is currently recognized as a negative regulator of muscle mass through inhibition of the Akt/mTORC1 signaling pathway. REDD1 expression is notably induced following glucocorticoid secretion, which is a component of energy stress responses. RESULTS: Unexpectedly, we show here that REDD1 instead limits muscle loss during energetic stresses such as hypoxia and fasting by reducing glycogen depletion and AMPK activation. Indeed, we demonstrate that REDD1 is required to decrease O2 and ATP consumption in skeletal muscle via reduction of the extent of mitochondrial-associated endoplasmic reticulum membranes (MAMs), a central hub connecting energy production by mitochondria and anabolic processes. In fact, REDD1 inhibits ATP-demanding processes such as glycogen storage and protein synthesis through disruption of the Akt/Hexokinase II and PRAS40/mTORC1 signaling pathways in MAMs. Our results uncover a new REDD1-dependent mechanism coupling mitochondrial respiration and anabolic processes during hypoxia, fasting, and exercise. CONCLUSIONS: Therefore, REDD1 is a crucial negative regulator of energy expenditure that is necessary for muscle adaptation during energetic stresses. This present study could shed new light on the role of REDD1 in several pathologies associated with energetic metabolism alteration, such as cancer, diabetes, and Parkinson's disease.


Assuntos
Metabolismo Energético/genética , Mitocôndrias Musculares/metabolismo , Músculo Esquelético/metabolismo , Estresse Fisiológico/genética , Fatores de Transcrição/fisiologia , Adaptação Fisiológica/genética , Animais , Hipóxia Celular , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Atrofia Muscular/genética , Transdução de Sinais , Fatores de Transcrição/genética
3.
Biochim Biophys Acta ; 1856(1): 144-50, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26116758

RESUMO

RIP140 is a transcriptional coregulator (also known as NRIP1) which plays very important physiological roles by finely tuning the activity of a large number of transcription factors. Noticeably, the RIP140 gene has been shown to be involved in the regulation of energy expenditure, in mammary gland development and intestinal homeostasis as well as in behavior and cognition. RIP140 is also involved in the regulation of various oncogenic signaling pathways and participates in the development and progression of solid tumors. This short review aims to summarize the role of this transcription factor on nuclear estrogen receptors, E2F and Wnt signaling pathways based on recent observations focusing on breast, ovary, liver and colon tumors.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Neoplasias/metabolismo , Proteínas Nucleares/fisiologia , Transcrição Gênica , Estrogênios/metabolismo , Feminino , Humanos , Masculino , Neoplasias/classificação , Proteína 1 de Interação com Receptor Nuclear , Transdução de Sinais , Proteínas Wnt/metabolismo
4.
Am J Physiol Endocrinol Metab ; 307(11): E983-93, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25315696

RESUMO

REDD1 (regulated in development and DNA damage response 1) has been proposed to inhibit the mechanistic target of rapamycin complex 1 (mTORC1) during in vitro hypoxia. REDD1 expression is low under basal conditions but is highly increased in response to several catabolic stresses, like hypoxia and glucocorticoids. However, REDD1 function seems to be tissue and stress dependent, and its role in skeletal muscle in vivo has been poorly characterized. Here, we investigated the effect of REDD1 deletion on skeletal muscle mass, protein synthesis, proteolysis, and mTORC1 signaling pathway under basal conditions and after glucocorticoid administration. Whereas skeletal muscle mass and typology were unchanged between wild-type (WT) and REDD1-null mice, oral gavage with dexamethasone (DEX) for 7 days reduced tibialis anterior and gastrocnemius muscle weights as well as tibialis anterior fiber size only in WT. Similarly, REDD1 deletion prevented the inhibition of protein synthesis and mTORC1 activity (assessed by S6, 4E-BP1, and ULK1 phosphorylation) observed in gastrocnemius muscle of WT mice following single DEX administration for 5 h. However, our results suggest that REDD1-mediated inhibition of mTORC1 in skeletal muscle is not related to the modulation of the binding between TSC2 and 14-3-3. In contrast, our data highlight a new mechanism involved in mTORC1 inhibition linking REDD1, Akt, and PRAS40. Altogether, these results demonstrated in vivo that REDD1 is required for glucocorticoid-induced inhibition of protein synthesis via mTORC1 downregulation. Inhibition of REDD1 may thus be a strategy to limit muscle loss in glucocorticoid-mediated atrophy.


Assuntos
Dexametasona , Atrofia Muscular/induzido quimicamente , Atrofia Muscular/genética , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia , Animais , Corticosterona/metabolismo , Fezes/química , Feminino , Camundongos , Contração Muscular/fisiologia , Músculo Esquelético/patologia , Atrofia Muscular/patologia , Proteólise , RNA Mensageiro/biossíntese , RNA Mensageiro/genética
5.
Front Immunol ; 11: 718, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32373132

RESUMO

Among inflammatory mediators, a growing body of evidence emphasizes the contribution of the interleukin 17 (IL-17) cytokine family in malignant diseases. Besides IL-17A, the prototypic member of the IL-17 family, several experimental findings strongly support the role of the IL-17B/IL-17 receptor B (IL-17RB) pathway in tumorigenesis and resistance to anticancer therapies. In mouse models, IL-17B signaling through IL-17RB directly promotes cancer cell survival, proliferation, and migration, and induces resistance to conventional chemotherapeutic agents. Importantly, recent work by our and other laboratories showed that IL-17B signaling dramatically alters the tumor microenvironment by promoting chemokine and cytokine secretion which foster tumor progression. Moreover, the finding that elevated IL-17B is associated with poor prognosis in patients with pancreatic, gastric, lung, and breast cancer strengthens the results obtained in pre-clinical studies and highlights its clinical relevance. Here, we review the current understanding on the IL-17B/IL-17RB expression patterns and biological activities in cancer and highlight issues that remain to be addressed to better characterize IL-17B and its receptor as potential targets for enhancing the effectiveness of the existing cancer therapies.


Assuntos
Interleucina-17/metabolismo , Neoplasias/imunologia , Receptores de Interleucina-17/metabolismo , Animais , Antineoplásicos/farmacologia , Modelos Animais de Doenças , Humanos , Camundongos , Terapia de Alvo Molecular/métodos , Neoplasias/tratamento farmacológico , Prognóstico , Transdução de Sinais/efeitos dos fármacos
6.
Cell Rep ; 27(8): 2411-2425.e9, 2019 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-31116985

RESUMO

Immune checkpoint inhibitors have revolutionized cancer treatment. However, many cancers are resistant to ICIs, and the targeting of additional inhibitory signals is crucial for limiting tumor evasion. The production of adenosine via the sequential activity of CD39 and CD73 ectoenzymes participates to the generation of an immunosuppressive tumor microenvironment. In order to disrupt the adenosine pathway, we generated two antibodies, IPH5201 and IPH5301, targeting human membrane-associated and soluble forms of CD39 and CD73, respectively, and efficiently blocking the hydrolysis of immunogenic ATP into immunosuppressive adenosine. These antibodies promoted antitumor immunity by stimulating dendritic cells and macrophages and by restoring the activation of T cells isolated from cancer patients. In a human CD39 knockin mouse preclinical model, IPH5201 increased the anti-tumor activity of the ATP-inducing chemotherapeutic drug oxaliplatin. These results support the use of anti-CD39 and anti-CD73 monoclonal antibodies and their combination with immune checkpoint inhibitors and chemotherapies in cancer.


Assuntos
5'-Nucleotidase/imunologia , Anticorpos Bloqueadores/imunologia , Antígenos CD/imunologia , Apirase/imunologia , 5'-Nucleotidase/genética , 5'-Nucleotidase/metabolismo , Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Anticorpos Bloqueadores/uso terapêutico , Antígenos CD/genética , Antineoplásicos/uso terapêutico , Apirase/deficiência , Apirase/genética , Linhagem Celular Tumoral , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Humanos , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/metabolismo , Melanoma/tratamento farmacológico , Melanoma/imunologia , Melanoma/mortalidade , Melanoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxaliplatina/uso terapêutico , Taxa de Sobrevida , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Microambiente Tumoral
7.
Int J Biochem Cell Biol ; 45(10): 2158-62, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23769948

RESUMO

The eukaryotic initiation factor 3 subunit f (eIF3f) is one of the 13 subunits of the translation initiation factor complex eIF3 required for several steps in the initiation of mRNA translation. In skeletal muscle, recent studies have demonstrated that eIF3f plays a central role in skeletal muscle size maintenance. Accordingly, eIF3f overexpression results in hypertrophy through modulation of protein synthesis via the mTORC1 pathway. Importantly, eIF3f was described as a target of the E3 ubiquitin ligase MAFbx/atrogin-1 for proteasome-mediated breakdown under atrophic conditions. The biological importance of the MAFbx/atrogin-1-dependent targeting of eFI3f is highlighted by the finding that expression of an eIF3f mutant insensitive to MAFbx/atrogin-1 polyubiquitination is associated with enhanced protection against starvation-induced muscle atrophy. A better understanding of the precise role of this subunit should lead to the development of new therapeutic approaches to prevent or limit muscle wasting that prevails in numerous physiological and pathological states such as immobilization, aging, denervated conditions, neuromuscular diseases, AIDS, cancer, diabetes. This article is part of a Directed Issue entitled: Molecular basis of muscle wasting.


Assuntos
Fator de Iniciação 3 em Eucariotos/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/patologia , Atrofia Muscular/metabolismo , Animais , Proliferação de Células , Fator de Iniciação 3 em Eucariotos/genética , Humanos , Proteínas Musculares/genética , Músculo Esquelético/metabolismo , Atrofia Muscular/genética , Atrofia Muscular/patologia , Biossíntese de Proteínas , Transdução de Sinais
8.
Mol Endocrinol ; 27(9): 1429-41, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23885094

RESUMO

In hormone-dependent tissues such as breast and ovary, tumorigenesis is associated with an altered expression ratio between the two estrogen receptor (ER) subtypes. In this study, we investigated the effects of ERß ectopic expression on 17ß-estradiol (E2)-induced transactivation and cell proliferation in ERα-positive BG1 ovarian cancer cells. As expected, ERß expression strongly decreased the mitogenic effect of E2, significantly reduced E2-dependent transcriptional responses (both on a stably integrated estrogen response element [ERE] reporter gene and on E2-induced mRNAs), and strongly enhanced the formation of ER heterodimers as evidenced by chromatin immunoprecipitation analysis. Inhibition by the ERα-selective ligand propyl pyrazole triol was less marked than with the pan-agonist (E2) or the ERß-selective (8ß-vinyl-estradiol) ligands, indicating that ERß activation reinforced the inhibitory effects of ERß. Interestingly, in E2-stimulated BG1 cells, ERß was more efficient than ERα to regulate the expression of receptor-interacting protein 140 (RIP140), a major ERα transcriptional corepressor. In addition, we found that the RIP140 protein interacted better with ERß than with ERα (both in vitro and in intact cells by fluorescence cross-correlation spectroscopy). Moreover, RIP140 recruitment on the stably integrated reporter ERE was increased upon ERß overexpression, and ERß activity was more sensitive to repression by RIP140. Finally, small interfering RNA-mediated knockdown of RIP140 expression abolished the repressive effect exerted by activated ERß on the regulation of ERE-controlled transcription by estrogens. Altogether, these data demonstrate the inhibitory effects of ERß on estrogen signaling in ovarian cancer cells and the key role that RIP140 plays in this phenomenon.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Receptor beta de Estrogênio/metabolismo , Estrogênios/metabolismo , Proteínas Nucleares/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Transdução de Sinais , Animais , Células COS , Linhagem Celular Tumoral , Proliferação de Células , Chlorocebus aethiops , DNA de Neoplasias/metabolismo , Receptor alfa de Estrogênio/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Genes Reporter , Humanos , Ligantes , Proteína 1 de Interação com Receptor Nuclear , Neoplasias Ovarianas/genética , Ligação Proteica/genética , Ativação Transcricional/genética
9.
J Steroid Biochem Mol Biol ; 132(1-2): 176-85, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22652558

RESUMO

Most ovarian cancers are estrogen-positive and hormonal treatments using anti-estrogens or aromatase inhibitors are under investigation for treating the tumors that are resistant to conventional therapies. In this study, the long-term effects of two anti-estrogens, namely 4-hydroxytamoxifen and fulvestrant (or ICI182,780), were investigated in ERα-positive BG1 epithelial ovarian cancer cells. To this aim, cells were grown in the presence of anti-estrogen concentrations that were sufficient to saturate the estrogen receptors, but were neither cytotoxic nor cytostatic as indicated by the absence of inhibition of cell proliferation. In these conditions and despite the lack of cytostatic effect of the drugs, long-term treatment (3 months) with the pure anti-estrogen fulvestrant induced a specific, reproducible and irreversible inhibition of ERα expression. This inhibition was accompanied by loss of estrogen-induced cell proliferation and gene expression as indicated by the analysis of several estrogen-responsive genes. ERα down-regulation was not linked to deregulated expression of transcription factors which drive ERα transcription and did not involve DNA methylation or histone deacetylation. Altogether, these results demonstrate that non-cytotoxic concentrations of pure anti-estrogens affect estrogen signaling and might be relevant for the treatment for ovarian cancers.


Assuntos
Estradiol/análogos & derivados , Antagonistas de Estrogênios/farmacologia , Estrogênios/metabolismo , Neoplasias Ovarianas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Estradiol/farmacologia , Receptor alfa de Estrogênio/antagonistas & inibidores , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Fulvestranto , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacologia
10.
PLoS One ; 7(5): e35839, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22629304

RESUMO

RIP140 is a transcriptional coregulator involved in energy homeostasis and ovulation which is controlled at the transcriptional level by several nuclear receptors. We demonstrate here that RIP140 is a novel target gene of the E2F1 transcription factor. Bioinformatics analysis, gel shift assay, and chromatin immunoprecipitation demonstrate that the RIP140 promoter contains bona fide E2F response elements. In transiently transfected MCF-7 breast cancer cells, the RIP140 promoter is transactivated by overexpression of E2F1/DP1. Interestingly, RIP140 mRNA is finely regulated during cell cycle progression (5-fold increase at the G1/S and G2/M transitions). The positive regulation by E2F1 requires sequences located in the proximal region of the promoter (-73/+167), involves Sp1 transcription factors, and undergoes a negative feedback control by RIP140. Finally, we show that E2F1 participates in the induction of RIP140 expression during adipocyte differentiation. Altogether, this work identifies the RIP140 gene as a new transcriptional target of E2F1 which may explain some of the effect of E2F1 in both cancer and metabolic diseases.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Fator de Transcrição E2F1/genética , Proteínas Nucleares/genética , Linhagem Celular Tumoral , Células Cultivadas , Células HeLa , Humanos , Proteína 1 de Interação com Receptor Nuclear , Regiões Promotoras Genéticas
11.
Bull Cancer ; 98(10): 1201-8, 2011 Oct.
Artigo em Francês | MEDLINE | ID: mdl-22001823

RESUMO

Vitamin B6 is well-known for its role as a cofactor in many enzymatic reactions and recently, several epidemiological studies have highlighted the importance of this vitamin as a protective agent against various cancers: elevated vitamin B6 plasma levels were associated with a lower risk of colorectal cancer development, for example. In vivo studies have shown that vitamin B6 decreased cell proliferation and enhanced the immune response. At the cellular level, antioxidant, pro-apoptotic and anti-angiogenic effects have been identified. At the molecular level, vitamin B6 is able to inhibit the transactivation potential of various nuclear receptors. Interestingly, a recent paper has described the conjugation of vitamin B6 to RIP140 (receptor interacting protein of 140 kDa), a protein that acts as a transcriptional corepressor of nuclear receptors. This post-translational modification increases the transcriptional repression of RIP140 and regulates its subcellular localization and its ability to interact with different protein partners. Finally, vitamin B6 is involved in the methyl donor cycle ant thus, some of the antitumor properties of vitamin B6 may involve an indirect effect on the level of DNA or histone methylation. All of these mechanistic and clinical data justify further studies to decipher the mechanism of action of vitamin B6 and its clinical interest in combination with molecules typically used in chemotherapy or hormonal therapy.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/prevenção & controle , Proteínas Nucleares/metabolismo , Vitamina B 6/fisiologia , Animais , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/prevenção & controle , Metilação de DNA/fisiologia , Epigênese Genética/fisiologia , Histonas/metabolismo , Humanos , Neoplasias/metabolismo , Proteína 1 de Interação com Receptor Nuclear , Processamento de Proteína Pós-Traducional , Ratos , Receptores Citoplasmáticos e Nucleares/metabolismo
12.
Clin Cancer Res ; 16(11): 2959-70, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20410059

RESUMO

PURPOSE: Receptor-interacting protein of 140 kDa (RIP140) is a transcriptional cofactor for nuclear receptors involved in reproduction and energy homeostasis. Our aim was to investigate its role in the regulation of E2F1 activity and target genes both in breast cancer cell lines and in tumor biopsies. EXPERIMENTAL DESIGN: Glutathione S-transferase pull-down assays, coimmunoprecipitation experiments, and chromatin immunoprecipitation analysis were used to evidence interaction between RIP140 and E2F1. The effects of RIP140 expression on E2F1 activity were determined using transient transfection and quantification of E2F target mRNAs by quantitative real-time PCR. The effect on cell cycle was assessed by fluorescence-activated cell sorting analysis on cells overexpressing green fluorescent protein-tagged RIP140. A tumor microarray data set was used to investigate the expression of RIP140 and E2F1 target genes in 170 breast cancer patients. RESULTS: We first evidenced the complex interaction between RIP140 and E2F1 and showed that RIP140 represses E2F1 transactivation on various transiently transfected E2F target promoters and inhibits the expression of several E2F1 target genes (such as CCNE1 and CCNB2). In agreement with a role for RIP140 in the control of E2F activity, we show that increasing RIP140 levels results in a reduction in the proportion of cells in S phase in various human cell lines. Finally, analysis of human breast cancers shows that low RIP140 mRNA expression was associated with high E2F1 target gene levels and basal-like tumors. CONCLUSION: This study shows that RIP140 is a regulator of the E2F pathway, which discriminates luminal- and basal-like tumors, emphasizing the importance of these regulations for a clinical cancer phenotype.


Assuntos
Neoplasias da Mama/genética , Fator de Transcrição E2F1/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica , Neoplasia de Células Basais/genética , Correpressor 1 de Receptor Nuclear/fisiologia , Neoplasias da Mama/patologia , Ciclo Celular , Linhagem Celular Tumoral , Feminino , Humanos , Ativação Transcricional , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA