Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
PLoS Genet ; 19(10): e1011014, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37906604

RESUMO

Activating Transcription Factor 4 (ATF4) is an important regulator of gene expression in stress responses and developmental processes in many cell types. Here, we catalogued ATF4 binding sites in the human genome and identified overlaps with trait-associated genetic variants. We probed these genetic variants for allelic regulatory activity using a massively parallel reporter assay (MPRA) in HepG2 hepatoma cells exposed to tunicamycin to induce endoplasmic reticulum stress and ATF4 upregulation. The results revealed that in the majority of cases, the MPRA allelic activity of these SNPs was in agreement with the nucleotide preference seen in the ATF4 binding motif from ChIP-Seq. Luciferase and electrophoretic mobility shift assays in additional cellular models further confirmed ATF4-dependent regulatory effects for the SNPs rs532446 (GADD45A intronic; linked to hematological parameters), rs7011846 (LPL upstream; myocardial infarction), rs2718215 (diastolic blood pressure), rs281758 (psychiatric disorders) and rs6491544 (educational attainment). CRISPR-Cas9 disruption and/or deletion of the regulatory elements harboring rs532446 and rs7011846 led to the downregulation of GADD45A and LPL, respectively. Thus, these SNPs could represent examples of GWAS genetic variants that affect gene expression by altering ATF4-mediated transcriptional activation.


Assuntos
Fator 4 Ativador da Transcrição , Censos , Humanos , Fator 4 Ativador da Transcrição/genética , Sítios de Ligação/genética , Sequências Reguladoras de Ácido Nucleico , Estresse do Retículo Endoplasmático/genética
2.
Cancers (Basel) ; 13(10)2021 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-34066165

RESUMO

The proteasome is an appealing target for anticancer therapy and the proteasome inhibitor bortezomib has been approved for the treatment of several types of malignancies. However, the molecular mechanisms underlying cancer cell resistance to bortezomib remain poorly understood. In the current article, we investigate how modulation of the eIF2α-ATF4 stress pathway affects hepatoma cell response to bortezomib. Transcriptome profiling revealed that many ATF4 transcriptional target genes are among the most upregulated genes in bortezomib-treated HepG2 human hepatoma cells. While pharmacological enhancement of the eIF2α-ATF4 pathway activity results in the elevation of the activities of all branches of the unfolded protein response (UPR) and sensitizes cells to bortezomib toxicity, the suppression of ATF4 induction delays bortezomib-induced cell death. The pseudokinase TRIB3, an inhibitor of ATF4, is expressed at a high basal level in hepatoma cells and is strongly upregulated in response to bortezomib. To map genome-wide chromatin binding loci of TRIB3 protein, we fused a Flag tag to endogenous TRIB3 in HepG2 cells and performed ChIP-Seq. The results demonstrate that TRIB3 predominantly colocalizes with ATF4 on chromatin and binds to genomic regions containing the C/EBP-ATF motif. Bortezomib treatment leads to a robust enrichment of TRIB3 binding near genes induced by bortezomib and involved in the ER stress response and cell death. Disruption of TRIB3 increases C/EBP-ATF-driven transcription, augments ER stress and cell death upon exposure to bortezomib, while TRIB3 overexpression enhances cell survival. Thus, TRIB3, colocalizing with ATF4 and limiting its transcriptional activity, functions as a factor increasing resistance to bortezomib, while pharmacological over-activation of eIF2α-ATF4 can overcome the endogenous restraint mechanisms and sensitize cells to bortezomib.

3.
PLoS Pathog ; 13(9): e1006589, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28953979

RESUMO

The ability of intracellular pathogens to manipulate host-cell viability is critical to successful infection. Some pathogens promote host-cell survival to protect their replicative niche, whereas others trigger host-cell death to facilitate release and dissemination of the pathogen after intracellular replication has occurred. We previously showed that the intracellular fungal pathogen Histoplasma capsulatum (Hc) uses the secreted protein Cbp1 to actively induce apoptosis in macrophages; interestingly, cbp1 mutant strains are unable to kill macrophages and display severely reduced virulence in the mouse model of Hc infection. To elucidate the mechanism of Cbp1-induced host-cell death, we performed a comprehensive alanine scanning mutagenesis and identified all amino acid residues that are required for Cbp1 to trigger macrophage lysis. Here we demonstrate that Hc strains expressing lytic CBP1 alleles activate the integrated stress response (ISR) in infected macrophages, as indicated by an increase in eIF2α phosphorylation as well as induction of the transcription factor CHOP and the pseudokinase Tribbles 3 (TRIB3). In contrast, strains bearing a non-lytic allele of CBP1 fail to activate the ISR, whereas a partially lytic CBP1 allele triggers intermediate levels of activation. We further show that macrophages deficient for CHOP or TRIB3 are partially resistant to lysis during Hc infection, indicating that the ISR is critical for susceptibility to Hc-mediated cell death. Moreover, we show that CHOP-dependent macrophage lysis is critical for efficient spread of Hc infection to other macrophages. Notably, CHOP knockout mice display reduced macrophage apoptosis and diminished fungal burden and are markedly resistant to Hc infection. Together, these data indicate that Cbp1 is required for Hc to induce the ISR and mediate a CHOP-dependent virulence pathway in the host.


Assuntos
Apoptose/imunologia , Genes Fúngicos/genética , Histoplasma/metabolismo , Histoplasmose/microbiologia , Macrófagos/metabolismo , Fator de Transcrição CHOP/metabolismo , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Feminino , Interações Hospedeiro-Patógeno/imunologia , Macrófagos/microbiologia , Camundongos , Virulência/genética
4.
Sci Rep ; 7(1): 6684, 2017 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-28751721

RESUMO

Megakaryocytopoiesis is a complex differentiation process driven by the hormone thrombopoietin by which haematopoietic progenitor cells give rise to megakaryocytes, the giant bone marrow cells that in turn break down to form blood platelets. The Tribbles Pseudokinase 3 gene (TRIB3) encodes a pleiotropic protein increasingly implicated in the regulation of cellular differentiation programmes. Previous studies have hinted that TRIB3 could be also involved in megakaryocytopoiesis but its role in this process has so far not been investigated. Using cellular model systems of haematopoietic lineage differentiation here we demonstrate that TRIB3 is a negative modulator of megakaryocytopoiesis. We found that in primary cultures derived from human haematopoietic progenitor cells, thrombopoietin-induced megakaryocytic differentiation led to a time and dose-dependent decrease in TRIB3 mRNA levels. In the haematopoietic cell line UT7/mpl, silencing of TRIB3 increased basal and thrombopoietin-stimulated megakaryocyte antigen expression, as well as basal levels of ERK1/2 phosphorylation. In primary haematopoietic cell cultures, silencing of TRIB3 facilitated megakaryocyte differentiation. In contrast, over-expression of TRIB3 in these cells inhibited the differentiation process. The in-vitro identification of TRIB3 as a negative regulator of megakaryocytopoiesis suggests that in-vivo this gene could be important for the regulation of platelet production.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Megacariócitos/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Repressoras/metabolismo , Trombopoese , Proteínas de Ciclo Celular/genética , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Inativação Gênica/efeitos dos fármacos , Humanos , Megacariócitos/citologia , Megacariócitos/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Repressoras/genética , Trombopoese/efeitos dos fármacos , Trombopoetina/farmacologia
5.
Curr Protein Pept Sci ; 18(8): 819-842, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28393700

RESUMO

Tribbles homolog 3 (TRIB3) is a mammalian gene that is upregulated in response to several types of cell death-inducing cellular stress. The TRIB3 protein is a pseudokinase, a protein kinase-like scaffold with impaired catalytic activity. However, research has revealed it to be prolific at forming protein- protein interactions. By binding to and regulating the activity of several key proteins, including the protein kinase Akt and transcription factors ATF4, CHOP and NF-κB, TRIB3 is at a junction of several signaling pathways. This review begins by providing insights into the characteristic protein structure and gene expression regulation mechanisms of TRIB3. Further, the diverse reported molecular roles of TRIB3 as a regulator of cell death, stress responses, inflammation, cell differentiation, protein degradation and other processes are discussed. Special attention is devoted to the involvement of TRIB3 in the pathogenesis of cancer and type 2 diabetes, two fields where TRIB3 has generated particular interest, as well as considerable debate, from a biomedical standpoint. Throughout, emphasis is placed on results obtained from animal models with altered TRIB3 expression (Trib3 knockout or overexpression mice), in order to provide insight into the contributions of TRIB3 to physiology and disease at the organism level.


Assuntos
Proteínas de Ciclo Celular/genética , Diabetes Mellitus Tipo 2/genética , Regulação da Expressão Gênica , Neoplasias/genética , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Repressoras/genética , Transdução de Sinais , Fator 4 Ativador da Transcrição/genética , Fator 4 Ativador da Transcrição/metabolismo , Animais , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Morte Celular , Diferenciação Celular , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Humanos , Camundongos , Camundongos Transgênicos , NF-kappa B/genética , NF-kappa B/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteólise , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Repressoras/química , Proteínas Repressoras/metabolismo , Fator de Transcrição CHOP/genética , Fator de Transcrição CHOP/metabolismo
6.
Biochim Biophys Acta ; 1863(11): 2668-2680, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27526673

RESUMO

Arsenic, a metalloid with cytotoxic and carcinogenic effects related to the disruption of glutathione homeostasis, induces the expression of ATF4, a central transcription factor in the cellular stress response. However, the interplay between factors downstream of ATF4 is incompletely understood. In this article, we investigate the role of Tribbles homolog 3 (TRIB3), a regulatory member of the ATF4 pathway, in determining cell sensitivity to arsenite. Our results show that arsenite potently upregulates Trib3 mRNA and protein in an ATF4-dependent manner in mouse embryonic fibroblasts. Trib3-deficient cells display increased susceptibility to arsenite-induced cell death, which is rescued by re-expressing TRIB3. In cells lacking TRIB3, arsenite stress leads to markedly elevated mRNA and protein levels of Chac1, a gene that encodes a glutathione-degrading enzyme and is not previously known to be repressed by TRIB3. Analysis of the Chac1 promoter identified two regulatory elements that additively mediate the induction of Chac1 by arsenite and ATF4, as well as the robust suppression of Chac1 by TRIB3. Crucially, Chac1 silencing enhances glutathione levels and eliminates the increased susceptibility of Trib3-deficient cells to arsenite stress. Moreover, Trib3-deficient cells demonstrate an increased rate of glutathione consumption, which is abolished by Chac1 knockdown. Taken together, these data indicate that excessive Chac1 expression is detrimental to arsenite-treated cell survival and that TRIB3 is critical for restraining the pro-death potential of Chac1 during arsenite stress, representing a novel mechanism of cell viability regulation that occurs within the ATF4 pathway.


Assuntos
Arsenitos/toxicidade , Proteínas de Ciclo Celular/metabolismo , Resistência a Medicamentos , Fibroblastos/efeitos dos fármacos , Glutationa/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Compostos de Sódio/toxicidade , Fator 4 Ativador da Transcrição/genética , Fator 4 Ativador da Transcrição/metabolismo , Animais , Sítios de Ligação , Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Proteínas de Ciclo Celular/deficiência , Proteínas de Ciclo Celular/genética , Linhagem Celular Transformada , Sobrevivência Celular/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Fibroblastos/enzimologia , Fibroblastos/patologia , Regulação Enzimológica da Expressão Gênica , Genótipo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos Knockout , Fenótipo , Regiões Promotoras Genéticas , Interferência de RNA , Fatores de Tempo , Transfecção , gama-Glutamilciclotransferase
7.
Biochim Biophys Acta ; 1853(10 Pt A): 2492-505, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26094770

RESUMO

Glucose deprivation occurs in several human diseases, including infarctions and solid tumors, and leads to cell death. In this article, we investigate the role of the pseudokinase Tribbles homolog 3 (TRIB3) in the cellular stress response to glucose starvation using cell lines derived from HEK293, which is highly glycolytic under standard conditions. Our results show that TRIB3 mRNA and protein levels are strongly upregulated in glucose-deprived cells via the induction of activating transcription factor 4 (ATF4) by the endoplasmic reticulum (ER) stress sensor kinase PERK. Cell survival in glucose-deficient conditions is enhanced by TRIB3 overexpression and reduced by TRIB3 knockdown. Genome-wide gene expression profiling uncovered approximately 40 glucose deprivation-responsive genes that are affected by TRIB3, including several genes involved in signaling processes and metabolism. Based on transcription factor motif analysis, the majority of TRIB3-downregulated genes are target genes of ATF4, which TRIB3 is known to inhibit. The gene most substantially upregulated by TRIB3 is insulin-like growth factor binding protein 2 (IGFBP2). IGFBP2 mRNA and protein levels are downregulated in cells subjected to glucose deprivation, and reduced IGFBP2 expression aggravates cell death during glucose deficiency, while overexpression of IGFBP2 prolongs cell survival. Moreover, IGFBP2 silencing abrogates the pro-survival effect of TRIB3. Since TRIB3 augments IGFBP2 expression in glucose-starved cells, the data indicate that IGFBP2 contributes to the attenuation of cell death by TRIB3. These results implicate TRIB3 and IGFBP2, both of which are known to be overexpressed in several types of cancers, as pro-survival modulators of cell viability in nutrient-deficient microenvironments.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Regulação Neoplásica da Expressão Gênica , Glucose/metabolismo , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/biossíntese , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Repressoras/metabolismo , Regulação para Cima , Fator 4 Ativador da Transcrição/genética , Fator 4 Ativador da Transcrição/metabolismo , Proteínas de Ciclo Celular/genética , Sobrevivência Celular/genética , Inativação Gênica , Glucose/genética , Células HEK293 , Humanos , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Proteínas de Neoplasias/genética , Neoplasias/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Proteínas Repressoras/genética , Microambiente Tumoral/genética , eIF-2 Quinase/genética , eIF-2 Quinase/metabolismo
8.
Cell Immunol ; 280(1): 68-75, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23261831

RESUMO

Mast cells are the principal effectors of IgE-mediated immune responses, including allergic reactions. Tribbles homolog 3 (Trib3) encodes a pseudokinase implicated in the cellular stress response and has been linked to inflammation in certain situations. Here we report the role of Trib3 in mouse bone marrow-derived mast cells (BMMCs). Our results show that Trib3 mRNA expression in BMMCs is positively regulated by the growth factor interleukin (IL)-3. BMMCs originating from Trib3 knockout mice demonstrate unaltered differentiation kinetics and cell surface expression of mast cell markers. When challenged with transient IL-3 deprivation, Trib3-deficient BMMCs display delayed recovery, and during prolonged IL-3 starvation, cell death is accelerated in Trib3-null cultures. IgE-dependent and pharmacologically induced degranulation is impaired in Trib3-deficient BMMCs, as is activation-induced cytokine mRNA expression. Thus, Trib3 contributes to the survival and activity of primary cultured mast cells, which suggests a role for Trib3 in the modulation of the immune response.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Interleucina-3/farmacologia , Mastócitos/efeitos dos fármacos , Animais , Células da Medula Óssea/citologia , Proteínas de Ciclo Celular/biossíntese , Proteínas de Ciclo Celular/deficiência , Proteínas de Ciclo Celular/genética , Degranulação Celular/efeitos dos fármacos , Degranulação Celular/fisiologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Fase G1/efeitos dos fármacos , Imunoglobulina E/imunologia , Interleucina-3/deficiência , Interleucina-3/fisiologia , Mastócitos/imunologia , Mastócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Anafilaxia Cutânea Passiva , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real
9.
PLoS One ; 5(12): e15716, 2010 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-21203563

RESUMO

In mammals, plasma amino acid concentrations are markedly affected by dietary or pathological conditions. It has been well established that amino acids are involved in the control of gene expression. Up to now, all the information concerning the molecular mechanisms involved in the regulation of gene transcription by amino acid availability has been obtained in cultured cell lines. The present study aims to investigate the mechanisms involved in transcriptional activation of the TRB3 gene following amino acid limitation in mice liver. The results show that TRB3 is up-regulated in the liver of mice fed a leucine-deficient diet and that this induction is quickly reversible. Using transient transfection and chromatin immunoprecipitation approaches in hepatoma cells, we report the characterization of a functional Amino Acid Response Element (AARE) in the TRB3 promoter and the binding of ATF4, ATF2 and C/EBPß to this AARE sequence. We also provide evidence that only the binding of ATF4 to the AARE plays a crucial role in the amino acid-regulated transcription of TRB3. In mouse liver, we demonstrate that the GCN2/eIF2α/ATF4 pathway is essential for the induction of the TRB3 gene transcription in response to a leucine-deficient diet. Therefore, this work establishes for the first time that the molecular mechanisms involved in the regulation of gene transcription by amino acid availability are functional in mouse liver.


Assuntos
Fator 4 Ativador da Transcrição/metabolismo , Proteínas de Ciclo Celular/metabolismo , Fator de Iniciação 2 em Eucariotos/metabolismo , Regulação Neoplásica da Expressão Gênica , Regulação da Expressão Gênica , Fígado/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Repressoras/metabolismo , Animais , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Humanos , Leucina/química , Camundongos , Elementos de Resposta
10.
Gene ; 444(1-2): 24-32, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19505541

RESUMO

Tribbles homolog 3 (TRB3) is a pseudokinase that has been implicated in the control of stress response, cell viability and metabolic processes, and has been linked to medical conditions, including insulin resistance, cardiovascular disease and diabetes. Therefore, the understanding of mechanisms that regulate TRB3 expression is of considerable importance. We have previously described the existence of several human (h) TRB3 mRNA isoforms that differ in their 5'-untranslated region (5'-UTR). In this study, we use a reverse transcription-quantitative polymerase chain reaction (RT-qPCR) system to characterize the expression levels of hTRB3 mRNA isoforms in HepG2 hepatoma cells cultured in regular medium or exposed to arsenite, and investigate the effect of hTRB3 5'-UTR variants on the efficiency of mRNA translation. The data indicate that of the hTRB3 mRNA splice variants, 1A is predominant (>80% of molecules) in both the stressed and unstressed states, and that the remainder consists mainly of 1B4, with the variants 1B1, 1B2 and 1B3 together forming less than 1% of the population in either condition. In addition to the substantial transcriptional upregulation of all hTRB3 mRNA splice variants, the exposure of cells to arsenite results in a marked increase in the proportion of splice variant 1A molecules containing a truncated 5'-UTR. The shortened 1A 5'-UTR proved to be translationally more efficient than the untruncated 1A 5'-UTR, due to the lack of an inhibitory upstream open reading frame (uORF). Thus, increased transcription as well as altered usage of 5'-UTR variants contributes to the upregulation of hTRB3 protein synthesis in stressful conditions.


Assuntos
Proteínas de Ciclo Celular/biossíntese , Proteínas Serina-Treonina Quinases/biossíntese , RNA Mensageiro/biossíntese , Proteínas Repressoras/biossíntese , Regiões 5' não Traduzidas , Arsenitos/farmacologia , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Humanos , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/genética , Proteínas Serina-Treonina Quinases/genética , Splicing de RNA , Proteínas Repressoras/genética , Estresse Fisiológico , Transcrição Gênica , Regulação para Cima
11.
Exp Cell Res ; 313(16): 3556-67, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17707795

RESUMO

Tribbles homolog 3 (TRB3) is a pseudokinase the level of which is increased in response to various stresses. We and other researchers have previously shown that TRB3 interacts with activating transcription factor 4 (ATF4) and may function as a negative feedback regulator of ATF4. In the present study, we investigate the effect of ATF4 and TRB3 on cell growth and viability, using both the enforced expression and silencing of the genes. HEK293 cells overexpressing ATF4 show retarded growth in the complete medium and decreased viability in the glucose-free medium. The enforced expression of ATF4 increases the level of reactive oxygen species (ROS) and the supplementation of the medium with ROS scavenging and reducing compounds supports the growth and survival of cells overexpressing ATF4. The deleterious effects of elevated ATF4 are suppressed by the coexpression of TRB3, which downregulates ATF4 transcriptional activity and results in the decrease of intracellular ROS. Also, the coexpression of TRB3 rescues postmitotic neuronally differentiated PC12 cells from the apoptosis evoked by ATF4 overexpression. The silencing of ATF4 and TRB3 genes by RNA interference reveals that endogenous ATF4 promotes and TRB3 suppresses the death of glucose-deprived SaOS2 cells. Together, the results indicate that TRB3 protects cells against the growth inhibitory and cytotoxic effect of ATF4.


Assuntos
Fator 4 Ativador da Transcrição/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Repressoras/metabolismo , Animais , Apoptose , Morte Celular , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células , Glucose/deficiência , Humanos , Células PC12 , Ratos , Espécies Reativas de Oxigênio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA