Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Biomolecules ; 11(8)2021 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-34439745

RESUMO

CCAAT/enhancer-binding protein delta (C/EBPδ) is a transcription factor involved in apoptosis and proliferation, which is downregulated in pancreatic ductal adenocarcinoma (PDAC) cells. Loss of nuclear C/EBPδ in PDAC cells is associated with decreased patient survival and pro-tumorigenic properties in vitro. Interestingly however, next to C/EBPδ expression in tumor cells, C/EBPδ is also expressed by cells constituting the tumor microenvironment and by cells comprising the organs and parenchyma. However, the functional relevance of systemic C/EBPδ in carcinogenesis remains elusive. Here, we consequently assessed the potential importance of C/EBPδ in somatic tissues by utilizing an orthotopic pancreatic cancer model. In doing so, we show that genetic ablation of C/EBPδ does not significantly affect primary tumor growth but has a strong impact on metastases; wildtype mice developed metastases at multiple sites, whilst this was not the case in C/EBPδ-/- mice. In line with reduced metastasis formation in C/EBPδ-/- mice, C/EBPδ-deficiency also limited tumor cell dissemination in a specific extravasation model. Tumor cell extravasation was dependent on the platelet-activating factor receptor (PAFR) as a PAFR antagonist inhibited tumor cell extravasation in wildtype mice but not in C/EBPδ-/- mice. Overall, we show that systemic C/EBPδ facilitates pancreatic cancer metastasis, and we suggest this is due to C/EBPδ-PAFR-dependent tumor cell extravasation.


Assuntos
Proteína delta de Ligação ao Facilitador CCAAT/biossíntese , Metástase Neoplásica , Neoplasias Pancreáticas/metabolismo , Animais , Apoptose , Proteína delta de Ligação ao Facilitador CCAAT/genética , Linhagem Celular Tumoral , Proliferação de Células , Proteínas de Ligação a DNA/metabolismo , Mineração de Dados , Progressão da Doença , Regulação da Expressão Gênica , Humanos , Camundongos , Transplante de Neoplasias , Neoplasias Pancreáticas/patologia , Glicoproteínas da Membrana de Plaquetas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Análise Serial de Tecidos , Transativadores/metabolismo , Microambiente Tumoral
2.
Biochem J ; 474(20): 3421-3437, 2017 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-28887384

RESUMO

Adipogenic differentiation plays a vital role in energy homeostasis and endocrine system. Several transcription factors, including peroxisome proliferator-activated receptor gamma 2 and CCAAT-enhancer-binding protein (C/EBP) α, ß, and δ, are important for the process, whereas the stage-specific intracellular signal transduction regulating the onset of adipogenesis remains enigmatic. Here, we explored the functional role of c-jun N-terminal kinases (JNKs) in adipogenic differentiation using in vitro differentiation models of 3T3-L1 cells and primary adipo-progenitor cells. JNK inactivation with either a pharmacological inhibitor or JNK2-specific siRNA suppressed adipogenic differentiation, characterized by decreased lipid droplet appearance and the down-regulation of Adiponectin, fatty acid protein 4 (Fabp4), Pparg2, and C/ebpa expressions. Conversely, increased adipogenesis was observed by the inducible overexpression of p46JNK2 (JNK2-1), whereas it was not observed by that of p54JNK2 (JNK2-2), indicating a distinct role of p46JNK2. The essential role of JNK appears restricted to the early stage of adipogenic differentiation, as JNK inhibition in the later stages did not influence adipogenesis. Indeed, JNK phosphorylation was significantly induced at the onset of adipogenic differentiation. As for the transcription factors involved in early adipogenesis, JNK inactivation significantly inhibited the induction of C/ebpd, but not C/ebpb, during the initial stage of adipogenic differentiation. JNK activation increased C/ebpd mRNA and protein expression through the induction and phosphorylation of activating transcription factor 2 (ATF2) that binds to a responsive element within the C/ebpd gene promoter region. Taken together, these data indicate that constitutive JNK activity is specifically required for the initial stage differentiation events of adipocytes.


Assuntos
Adipogenia/fisiologia , Proteína delta de Ligação ao Facilitador CCAAT/biossíntese , Diferenciação Celular/fisiologia , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Células 3T3-L1 , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Adipogenia/efeitos dos fármacos , Animais , Antracenos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 9 Ativada por Mitógeno/antagonistas & inibidores
3.
Gene ; 539(1): 141-51, 2014 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-24440782

RESUMO

Bone cells respond to the integrated effects of local and systemic regulation. Here we show that hypoxia and the stress hormones PGE2 and glucocorticoid interact in complex ways in osteoblasts, converging on insulin like growth factor I (IGF-I) expression. Whereas hypoxia alone rapidly increased transcription factor HIF activity, it suppressed DNA synthesis, had no significant effects on protein synthesis or alkaline phosphatase activity, and drove discrete changes in a panel of osteoblast mRNAs. Notably, hypoxia increased expression of the acute phase response transcription factor C/EBPδ which can induce IGF-I in response to PGE2, but conversely prevented the stimulatory effect of PGE2 on IGF-I mRNA. However, unlike its effect on C/EBPδ, hypoxia suppressed expression of the obligate osteoblast transcription factor Runx2, which can activate an upstream response element in the IGF-I gene promoter. Hypoxic inhibition of IGF-I and Runx2 were enforced by glucocorticoid, and continued with prolonged exposure. Our studies thus reveal that IGF-I expression is stratified by two critical transcriptional elements in osteoblasts, which are resolved by the individual and combined effects of hypoxic stress and stress hormones. In so doing, hypoxia suppresses Runx2, limits the enhancing influence of PGE2, and interacts with glucocorticoid to reduce IGF-I expression by osteoblasts.


Assuntos
Hipóxia Celular/genética , Dinoprostona/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Osteoblastos/metabolismo , Fosfatase Alcalina/metabolismo , Animais , Proteína delta de Ligação ao Facilitador CCAAT/biossíntese , Proteína delta de Ligação ao Facilitador CCAAT/genética , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/biossíntese , Subunidade alfa 1 de Fator de Ligação ao Core/genética , DNA/biossíntese , Replicação do DNA , Regulação da Expressão Gênica/genética , Hidrocortisona/metabolismo , Fator de Crescimento Insulin-Like I/biossíntese , Fator de Crescimento Insulin-Like I/genética , Osteoblastos/citologia , Oxigênio/metabolismo , Regiões Promotoras Genéticas , Biossíntese de Proteínas , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Elementos Reguladores de Transcrição/genética
4.
Int J Biochem Cell Biol ; 45(7): 1281-92, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23557604

RESUMO

ß-Arrestins were identified as scaffold-proteins that have the capacity to desensitize G protein-coupled receptors. However, it has been found that ß-arrestins activate signaling pathways independent of G protein activation. The diversity of these signaling pathways has also been recognized for receptor tyrosine kinase. The aim of the present study was to validate the ß-arrestin-dependent signaling mechanism(s) responsible for regulation of adipogenesis. Two signal models were selected, ghrelin and insulin, based on its ß-arrestin-associated Akt activity. Herein, we found that ß-arrestin 1 and 2 were essential molecules for adipocyte differentiation. More specifically, the role of these scaffolding proteins was demonstrated by depletion of ß-arrestin 1 and 2 during ghrelin-induced adipogenesis in 3T3-L1 cells, which decreased the adipocyte differentiation and the expression levels of master regulators of early, the CCAAT/enhancer-binding protein ß (C/EBPß) and the CCAAT/enhancer-binding protein δ (C/EBPδ), and terminal, the peroxisome proliferator-activated receptor (PPARγ) and the CCAAT/enhancer-binding protein α (C/EBPα), adipogenesis. Accordingly ghrelin-induced Akt activity and its downstream targets, the mammalian target of rapamycin complex 1 (mTORC1) and the ribosomal protein S6 kinase beta-1 (S6K1), were inhibited by ß-arrestin 1 and 2 siRNAs. By contrast, assays performed during insulin-activated adipogenesis showed an intensifying effect on the adipocyte differentiation as well as on the expression of C/EBPß, C/EBPδ, PPARγ and C/EBPα. The increase in insulin-induced adipogenesis by ß-arrestin knock-down was concomitant to a decrease in the insulin receptor susbtrate-1 (IRS-1) serine phosphorylation, proving the loss of the negative feedback loop on IRS-1/phosphoinositide 3-kinase (PI3K)/Akt. Therefore, ß-arrestins control the extent and intensity of the lipogenic and adipogenic factors associated to Akt signaling, although the mechanistic and functional principles that underlie the connection between signaling and ß-arrestins are specifically associated to each receptor type.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Arrestinas/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Células 3T3 , Adipócitos/citologia , Tecido Adiposo/citologia , Animais , Arrestinas/genética , Proteína beta Intensificadora de Ligação a CCAAT/biossíntese , Proteína delta de Ligação ao Facilitador CCAAT/biossíntese , Proteínas Estimuladoras de Ligação a CCAAT/biossíntese , Diferenciação Celular , Linhagem Celular , Grelina/metabolismo , Insulina/metabolismo , Camundongos , PPAR gama/biossíntese , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Transdução de Sinais , beta-Arrestina 1 , beta-Arrestinas
5.
J Cell Biochem ; 113(7): 2488-99, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22396222

RESUMO

Adipocyte dysfunction is associated with the development of obesity. In this study, artemisinic acid, which was isolated from Artemisia annua L., inhibited adipogenic differentiation of human adipose tissue-derived mesenchymal stem cells (hAMSCs) and its mechanism of action was determined. The mRNA levels of peroxidase proliferation-activated receptor (PPAR) γ and CCAAT/enhancer binding protein (C/EBP) α, late adipogenic factors, were reduced by artemisinic acid. Moreover, the mRNA levels of the PPAR γ target genes lipoprotein lipase, CD36, adipocyte protein, and liver X receptor were down-regulated by artemisinic acid. Artemisinic acid reduced expression of the C/EBP δ gene without impacting C/EBP ß. In addition, attempts to elucidate a possible mechanism underlying the artemisinic acid-mediated effects revealed that reduced expression of the C/EBP δ gene was mediated by inhibiting Jun N-terminal kinase (JNK). Additionally, artemisinic acid also reduced the expression of the adipogenesis-associated genes glucose transporter-4 and vascular endothelial growth factor. In addition to the interference of artemisinic acid with adipogenesis, artemisinic acid significantly attenuated tumor necrosis factor-α-induced secretion of interleukin-6 by undifferentiated hAMSCs, thus influencing insulin resistance and the inflammatory state characterizing obesity. Taken together, these findings indicate that inhibiting adipogenic differentiation of hAMSCs by artemisinic acid occurs primarily through reduced expression of C/EBP δ, which is mediated by the inhibition of JNK and suggest that aremisinic acid may be used as a complementary treatment option for obesity associated with metabolic syndrome.


Assuntos
Adipogenia/efeitos dos fármacos , Artemisininas/farmacologia , Proteína delta de Ligação ao Facilitador CCAAT/biossíntese , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Obesidade/tratamento farmacológico , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Adipócitos/fisiologia , Antígenos CD36/biossíntese , Antígenos CD36/genética , Diferenciação Celular , Linhagem Celular , Regulação para Baixo , Medicamentos de Ervas Chinesas/farmacologia , Proteínas de Ligação a Ácido Graxo/biossíntese , Proteínas de Ligação a Ácido Graxo/genética , Transportador de Glucose Tipo 4/biossíntese , Humanos , Resistência à Insulina , Interleucina-6/biossíntese , Interleucina-6/metabolismo , Lipase Lipoproteica/biossíntese , Lipase Lipoproteica/genética , Receptores X do Fígado , Células-Tronco Mesenquimais/fisiologia , Receptores Nucleares Órfãos/biossíntese , Receptores Nucleares Órfãos/genética , PPAR gama/biossíntese , PPAR gama/genética , RNA Mensageiro/biossíntese , Fator de Necrose Tumoral alfa/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese
6.
J Comp Physiol B ; 181(6): 773-80, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21442321

RESUMO

Members of the CCAAT/enhancer-binding protein (C/EBP) family of transcription factors have regulatory control over numerous processes related to cell fate determination, including differentiation, proliferation, cell cycle arrest and apoptosis. In mammals, abnormalities in the expression of some isoforms of C/EBPs are pathogenic and are implicated as being involved in myeloid leukemia and breast cancers. Next to nothing is known about their regulation, function or stress-responsiveness in poikilotherms. Here, both acute heat stress and thermal acclimation were demonstrated to induce the expression of one isoform, C/EBP-δ, in the liver, white muscle and gill of the eurythermal estuarine goby, Gillichthys mirabilis. The established role of C/EBP-δ in causing cell cycle arrest and/or promoting apoptosis in other vertebrates suggests that the heat-inducibility of this protein in poikilotherms may be part of the conserved cellular stress response with the hypothesized role of causing temporary cessation of cell growth and/or programmed cell death during bouts of environmental stress. The observed regulation of c/ebp-δ during hyperthermia represents a novel, heat-inducible signaling pathway in fishes.


Assuntos
Aclimatação/fisiologia , Proteína delta de Ligação ao Facilitador CCAAT/biossíntese , Estresse Fisiológico/fisiologia , Animais , Proteína delta de Ligação ao Facilitador CCAAT/fisiologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Brânquias/metabolismo , Temperatura Alta , Fígado/metabolismo , Músculos/metabolismo , Perciformes
7.
Biochem Pharmacol ; 80(3): 325-34, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20385105

RESUMO

Bladder cancer is the fourth most common type of cancer in men (ninth in women) in the United States. Cisplatin is an effective agent against the most common subtype, urothelial carcinoma. However, the development of chemotherapy resistance is a severe clinical problem for the successful treatment of this and other cancers. A better understanding of the cellular and molecular events in response to cisplatin treatment and the development of resistance are critical to improve the therapeutic options for patients. Here, we report that expression of the CCAAT/enhancer binding protein delta (CEBPD, C/EBPdelta, NF-IL6beta) is induced by cisplatin in the human bladder urothelial carcinoma NTUB1 cell line and is specifically elevated in a cisplatin resistant subline. Expression of CEBPD reduced cisplatin-induced reactive oxygen species (ROS) and apoptosis in NTUB1 cells by inducing the expression of Cu/Zn-superoxide dismutase (SOD1) via direct promoter transactivation. Several reports have implicated CEBPD as a tumor suppressor gene. This study reveals a novel role for CEBPD in conferring drug resistance, suggesting that it can also be pro-oncogenic. Furthermore, our data suggest that SOD inhibitors, which are already used as anti-angiogenic agents, may be suitable for combinatorial chemotherapy to prevent or treat cisplatin resistance in bladder and possibly other cancers.


Assuntos
Proteína delta de Ligação ao Facilitador CCAAT/fisiologia , Cisplatino/administração & dosagem , Sistemas de Liberação de Medicamentos , Superóxido Dismutase/biossíntese , Ativação Transcricional/fisiologia , Neoplasias Urológicas/metabolismo , Apoptose/genética , Proteína delta de Ligação ao Facilitador CCAAT/biossíntese , Proteína delta de Ligação ao Facilitador CCAAT/genética , Linhagem Celular Transformada , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos/métodos , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Superóxido Dismutase/genética , Superóxido Dismutase-1 , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/fisiologia , Ativação Transcricional/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia , Neoplasias Urológicas/tratamento farmacológico , Neoplasias Urológicas/genética
8.
Mol Cancer ; 9: 92, 2010 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-20426839

RESUMO

BACKGROUND: "Loss of function" alterations in CCAAT/Enhancer Binding Proteindelta (C/EBPdelta) have been reported in a number of human cancers including breast, prostate and cervical cancer, hepatocellular carcinoma and acute myeloid leukemia. C/EBPdelta gene transcription is induced during cellular quiescence and repressed during active cell cycle progression. C/EBPdelta exhibits tumor suppressor gene properties including reduced expression in cancer cell lines and tumors and promoter methylation silencing. We previously reported that C/EBPdelta expression is inversely correlated with c-Myc (Myc) expression. Aberrant Myc expression is common in cancer and transcriptional repression is a major mechanism of Myc oncogenesis. A number of tumor suppressor genes are targets of Myc transcriptional repression including C/EBPalpha, p15INK4, p21CIP1, p27KIP1 and p57KIP2. This study investigated the mechanisms underlying Myc repression of C/EBPdelta expression. RESULTS: Myc represses C/EBPdelta promoter activity in nontransformed mammary epithelial cells in a dose-dependent manner that requires Myc Box II, Basic Region and HLH/LZ domains. Chromatin Immunoprecipitation (ChIP) assays demonstrate that Myc, Miz1 and Max are associated with the C/EBPdelta promoter in proliferating cells, when C/EBPdelta expression is repressed. EMSAs demonstrate that Miz1 binds to a 30 bp region (-100 to -70) of the C/EBPdelta promoter which contains a putative transcription initiator (Inr) element. Miz1 functions exclusively as a repressor of C/EBPdelta promoter activity. Miz1 siRNA expression or expression of a Miz1 binding deficient Myc (MycV394D) construct reduces Myc repression of C/EBPdelta promoter activity. Max siRNA expression, or expression of a Myc construct lacking the HLH/LZ (Max interacting) region, also reduces Myc repression of C/EBPdelta promoter activity. Miz1 and Max siRNA treatments attenuate Myc repression of endogenous C/EBPdelta expression. Myc Box II interacting proteins RuvBl1 (Pontin, TIP49) and RuvBl2 (Reptin, TIP48) enhances Myc repression of C/EBPdelta promoter activity. CONCLUSION: Myc represses C/EBPdelta expression by associating with the C/EBPdelta proximal promoter as a transient component of a repressive complex that includes Max and Miz1. RuvBl1 and RuvBl2 enhance Myc repression of C/EBPdelta promoter activity. These results identify protein interactions that mediate Myc repression of C/EBPdelta, and possibly other tumor suppressor genes, and suggest new therapeutic targets to block Myc transcriptional repression and oncogenic function.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Proteína delta de Ligação ao Facilitador CCAAT/biossíntese , Regulação da Expressão Gênica/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Glândulas Mamárias Animais/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Repressoras/metabolismo , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Western Blotting , Proteína delta de Ligação ao Facilitador CCAAT/genética , Imunoprecipitação da Cromatina , Ensaio de Desvio de Mobilidade Eletroforética , Células Epiteliais , Feminino , Expressão Gênica , Imunoprecipitação , Fatores de Transcrição Kruppel-Like/genética , Camundongos , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-myc/genética , RNA Interferente Pequeno , Proteínas Repressoras/genética , Transfecção
9.
J Neurooncol ; 97(2): 233-40, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19806320

RESUMO

CCAAT/enhancer binding protein (C/EBP) delta is a transcription factor which has been demonstrated to mediate the growth arrest of mammary and prostate cancer cell lines. It is induced by several stimuli including inflammatory cytokines. In this study, C/EBPdelta immunohistochemical expression was assessed in 49 meningiomas of different histotype and grade and correlated with a variety of clinico-pathological data and with the overall and recurrence-free survival of the patients. Positive staining was observed in the nuclei of neoplastic cells in 22 out of the 49 cases analyzed. C/EBPdelta expression was significantly associated with a low histological grade and proliferation index, reflected by low Ki-67 labeling index (LI) and mitotic activity, and with the presence of intra-tumoral inflammatory infiltrate and the absence of necrosis. In addition, the absence of C/EBPdelta was significantly correlated with a shorter disease-free interval. Our findings suggest that C/EBPdelta expression may prevent the development of recurrences by inhibition of neoplastic growth in meningiomas. If further studies confirm its induction by inflammatory mediators, this might be exploited in novel therapies to prevent recurrences in meningiomas.


Assuntos
Proteína delta de Ligação ao Facilitador CCAAT/biossíntese , Neoplasias Meníngeas/metabolismo , Meningioma/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Proteína delta de Ligação ao Facilitador CCAAT/genética , Proliferação de Células , Feminino , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Masculino , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/patologia , Meningioma/genética , Meningioma/patologia , Pessoa de Meia-Idade , Adulto Jovem
10.
J Mol Cell Cardiol ; 46(3): 395-404, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19073192

RESUMO

During the transition from juvenile to adult life, the heart undergoes programmed remodeling at the levels of transcription and alternative splicing. Members of the CUG-BP and ETR-3-like factor (CELF) family have been implicated in driving developmental transitions in alternative splicing of cardiac transcripts during maturation of the heart. Here, we investigated the timing of the requirement for CELF activity in the postnatal heart using a previously described transgenic mouse model (MHC-CELFDelta). In MHC-CELFDelta mice, nuclear CELF activity has been disrupted specifically in the heart by cardiac-specific expression of a dominant negative CELF protein. Longitudinal analyses of two lines of MHC-CELFDelta mice with differing levels of dominant negative protein expression demonstrate that CELF splicing activity is required for healthy cardiac function during juvenile, but not adult, life. Cardiac function, chamber dilation, and heart size all recover with age in the mild line of MHC-CELFDelta mice without a loss of dominant negative protein expression or change in expression of endogenous CELF proteins or known CELF antagonists. This is the first example of a mouse model with genetically induced cardiomyopathy that spontaneously recovers without intervention. Our results suggest that CELF proteins are key players in the integrated gene expression program involved in postnatal cardiac remodeling and maturation.


Assuntos
Envelhecimento/metabolismo , Processamento Alternativo/fisiologia , Proteína delta de Ligação ao Facilitador CCAAT/biossíntese , Coração/crescimento & desenvolvimento , Proteínas Musculares/biossíntese , Miocárdio/metabolismo , Envelhecimento/genética , Animais , Proteína delta de Ligação ao Facilitador CCAAT/genética , Cardiomiopatias/genética , Cardiomiopatias/metabolismo , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Proteínas Musculares/genética , Tamanho do Órgão/fisiologia , Especificidade de Órgãos/fisiologia
11.
J Cell Biochem ; 103(5): 1657-69, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-17902160

RESUMO

The G(0) growth arrest (quiescent) state is highly conserved in evolution to promote survival under adverse environmental conditions. To maintain viability, G(0) growth arrested cells limit gene expression to essential growth control and pro-survival genes. CCAAT enhancer binding protein delta (C/EBPdelta), a member of the C/EBP family of nuclear proteins, is highly expressed in G(0) growth arrested mammary epithelial cells (MECs). Although C/EBPdelta gene transcription is elevated during G(0) growth arrest, C/EBPdelta mRNA and protein are relatively short lived, suggesting tight control of the cellular C/EBPdelta content in unstressed, quiescent cells. Treatment of G(0) growth arrested MECs with ultraviolet radiation (UVR) dramatically increases the C/EBPdelta mRNA half-life (approximately 4-fold) and protein content (approximately 3-fold). The mRNA stabilizing effects of UVR treatment are mediated by the C/EBPdelta mRNA 3'untranslated region, which contains an AU rich element. UVR increased p38 MAP kinase (MAPK) activation and SB203580, a p38 MAPK inhibitor, blocked UVR-induced C/EBPdelta mRNA stabilization. UVR increased the nuclear to cytoplasmic translocation of HuR, an ARE-binding protein that functions in mRNA stabilization. Finally, HuR siRNA treatment blocked UVR-induced stabilization of the C/EBPdelta and C/EBPbeta mRNAs but had no effect on C/EBPzeta (CHOP) mRNA stability. In summary, G(0) growth arrested MECs respond to UVR treatment by activating p38 MAPK, increasing HuR translocation and HuR/C/EBPdelta mRNA binding and stabilizing the C/EBPdelta mRNA. These results identify post-transcriptional stabilization of the C/EBPdelta mRNA as a mechanism to increase C/EBPdelta levels in the stress response of quiescent cells to UVR.


Assuntos
Proteína delta de Ligação ao Facilitador CCAAT/biossíntese , Células Epiteliais/metabolismo , Glândulas Mamárias Animais/metabolismo , Estabilidade de RNA/efeitos da radiação , Fase de Repouso do Ciclo Celular/efeitos da radiação , Raios Ultravioleta , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Regiões 3' não Traduzidas/metabolismo , Animais , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos da radiação , Meia-Vida , Imidazóis/farmacologia , Camundongos , Piridinas/farmacologia , Estabilidade de RNA/efeitos dos fármacos , Proteínas de Ligação a RNA/metabolismo , Fase de Repouso do Ciclo Celular/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
12.
Am J Respir Crit Care Med ; 176(6): 602-9, 2007 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-17600275

RESUMO

RATIONALE: A runaway inflammatory response to systemic infection or severe trauma is characterized by the activation of a diversity of pathways, ultimately resulting in the development of disseminated intravascular coagulation (DIC) and multiorgan failure. OBJECTIVES: Despite increased fundamental knowledge of the pathogenesis of DIC, the exact molecular mechanisms remain elusive. We aimed therefore to improve our understanding of the molecular pathways underlying endotoxin-induced DIC. METHODS: We performed large-scale gene expression profiling in the liver of mice during the onset of endotoxin-induced DIC. The relevance of an identified candidate gene involved in endotoxin-induced DIC was subsequently assessed in the generalized Shwartzman reaction. MEASUREMENTS AND MAIN RESULTS: Approximately 5% of over 20,000 genes were differentially regulated. In addition to well-established sepsis-associated genes, such as macrophage inflammatory protein 1, plasminogen activator inhibitor 1, CD14, and A20, we identified several novel candidates for inflammatory disease of which the transcription factor C/EBPdelta (CAAT/enhancer binding protein delta) was studied further. Induction of DIC in C/EBPdelta-deficient mice decreased endotoxin-induced systemic inflammation as compared with wild-type mice, as evident from decreased plasma levels of tumor necrosis factor-alpha and IL-6. In addition, C/EBPdelta deficiency partly protected against DIC-induced mortality. Interestingly, C/EBPdelta deficiency seemed mainly protective by improving renal function. This latter notion was confirmed in an experimental model of renal ischemia/reperfusion injury in which C/EBPdelta deficiency reduced ischemia/reperfusion-induced creatinine and urea levels. CONCLUSIONS: Our results endorse the usefulness of gene expression profiling in identifying novel mediators of DIC by showing that C/EBPdelta regulates specific pathologic features of this endotoxin-induced syndrome.


Assuntos
Proteína delta de Ligação ao Facilitador CCAAT/genética , Coagulação Intravascular Disseminada/genética , Expressão Gênica , RNA Mensageiro/genética , Animais , Proteína delta de Ligação ao Facilitador CCAAT/biossíntese , Modelos Animais de Doenças , Progressão da Doença , Coagulação Intravascular Disseminada/etiologia , Coagulação Intravascular Disseminada/metabolismo , Endotoxinas/toxicidade , Hibridização in Situ Fluorescente , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase , Índice de Gravidade de Doença
13.
Biochem Biophys Res Commun ; 356(3): 727-32, 2007 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-17382296

RESUMO

Gene regulation mediated by STAT factors has been implicated in cellular functions with relevance to a variety of processes. Particularly, STAT5 and STAT3 play a crucial role in mammary epithelium displaying reciprocal activation kinetics during pregnancy, lactation and involution. Here, we show that LIF treatment of mammary epithelial HC11 cells reduces the phosphorylation levels and transcriptional activity of p-STAT5 in correlation with STAT3 phosphorylation. We have also found that STAT5 activity is negatively modulated by this cytokine, both on a gene whose expression is induced, as well as on a promoter repressed by STAT5. Besides, our results show that lactogenic hormones increase LIF effect on gene induction without modifying STAT3 phosphorylation state. Our findings strongly suggest that there is crosstalk between STAT5 and STAT3 pathways that could modulate their ability to regulate gene expression.


Assuntos
Fator Inibidor de Leucemia/fisiologia , Fator de Transcrição STAT3/fisiologia , Fator de Transcrição STAT5/fisiologia , Animais , Proteína delta de Ligação ao Facilitador CCAAT/biossíntese , Dexametasona/farmacologia , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Insulina/farmacologia , Glândulas Mamárias Animais , Camundongos , Fosforilação/efeitos dos fármacos , Prolactina/farmacologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/biossíntese , Fator de Transcrição STAT3/metabolismo , Proteína bcl-X/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA