Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Ther Methods Clin Dev ; 24: 20-29, 2022 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-34977269

RESUMO

Extensive clinical data from liver-mediated gene therapy trials have shown that dose-dependent immune responses against the vector capsid may impair or even preclude transgene expression if not managed successfully with prompt immune suppression. The goal of this preclinical study was to generate an adeno-associated viral (AAV) vector capable of expressing therapeutic levels of B-domain deleted factor VIII (FVIII) at the lowest possible vector dose to minimize the potential Risk of a capsid-mediated immune response in the clinical setting. Here, we describe the studies that identified the investigational agent SPK-8011, currently being evaluated in a phase 1/2 study (NCT03003533) in individuals with hemophilia A. In particular, the potency of our second-generation expression cassettes was evaluated in mice and in non-human primates using two different bioengineered capsids (AAV-Spark100 and AAV-Spark200). At 2 weeks after gene transfer, primates transduced with 2 × 1012 vg/kg AAV-Spark100-FVIII or AAV-Spark200-FVIII expressed FVIII antigen levels of 13% ± 2% and 22% ± 6% of normal, respectively. Collectively, these preclinical results validate the feasibility of lowering the AAV capsid dose for a gene-based therapeutic approach for hemophilia A to a dose level orders of magnitude lower than the first-generation vectors in the clinic.

2.
Nat Commun ; 12(1): 6393, 2021 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-34737297

RESUMO

Pompe disease (PD) is a severe neuromuscular disorder caused by deficiency of the lysosomal enzyme acid alpha-glucosidase (GAA). PD is currently treated with enzyme replacement therapy (ERT) with intravenous infusions of recombinant human GAA (rhGAA). Although the introduction of ERT represents a breakthrough in the management of PD, the approach suffers from several shortcomings. Here, we developed a mouse model of PD to compare the efficacy of hepatic gene transfer with adeno-associated virus (AAV) vectors expressing secretable GAA with long-term ERT. Liver expression of GAA results in enhanced pharmacokinetics and uptake of the enzyme in peripheral tissues compared to ERT. Combination of gene transfer with pharmacological chaperones boosts GAA bioavailability, resulting in improved rescue of the PD phenotype. Scale-up of hepatic gene transfer to non-human primates also successfully results in enzyme secretion in blood and uptake in key target tissues, supporting the ongoing clinical translation of the approach.


Assuntos
Doença de Depósito de Glicogênio Tipo II/enzimologia , alfa-Glucosidases/metabolismo , Animais , Autofagia , Terapia de Reposição de Enzimas , Feminino , Doença de Depósito de Glicogênio Tipo II/terapia , Fígado/enzimologia , Masculino , Camundongos , alfa-Glucosidases/genética
3.
Nat Med ; 26(7): 1096-1101, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32483358

RESUMO

Neutralizing antibodies to adeno-associated virus (AAV) vectors are highly prevalent in humans1,2, and block liver transduction3-5 and vector readministration6; thus, they represent a major limitation to in vivo gene therapy. Strategies aimed at overcoming anti-AAV antibodies are being studied7, which often involve immunosuppression and are not efficient in removing pre-existing antibodies. Imlifidase (IdeS) is an endopeptidase able to degrade circulating IgG that is currently being tested in transplant patients8. Here, we studied if IdeS could eliminate anti-AAV antibodies in the context of gene therapy. We showed efficient cleavage of pooled human IgG (intravenous Ig) in vitro upon endopeptidase treatment. In mice passively immunized with intravenous Ig, IdeS administration decreased anti-AAV antibodies and enabled efficient liver gene transfer. The approach was scaled up to nonhuman primates, a natural host for wild-type AAV. IdeS treatment before AAV vector infusion was safe and resulted in enhanced liver transduction, even in the setting of vector readministration. Finally, IdeS reduced anti-AAV antibody levels from human plasma samples in vitro, including plasma from prospective gene therapy trial participants. These results provide a potential solution to overcome pre-existing antibodies to AAV-based gene therapy.


Assuntos
Anticorpos Neutralizantes/imunologia , Dependovirus/genética , Terapia Genética , Vetores Genéticos/efeitos adversos , Animais , Anticorpos Anti-Idiotípicos/genética , Anticorpos Anti-Idiotípicos/imunologia , Anticorpos Neutralizantes/genética , Anticorpos Antivirais/imunologia , Capsídeo/imunologia , Dependovirus/imunologia , Endopeptidases/imunologia , Vetores Genéticos/uso terapêutico , Humanos , Imunoglobulina G/farmacologia , Fígado/imunologia , Fígado/metabolismo , Camundongos
4.
Nat Commun ; 8(1): 549, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28916805

RESUMO

The histone deacetylase HDAC3 is a critical mediator of hepatic lipid metabolism, and liver-specific deletion of HDAC3 leads to fatty liver. To elucidate the underlying mechanism, here we report a method of cross-linking followed by mass spectrometry to define a high-confidence HDAC3 interactome in vivo that includes the canonical NCoR-HDAC3 complex as well as Prospero-related homeobox 1 protein (PROX1). HDAC3 and PROX1 co-localize extensively on the mouse liver genome, and are co-recruited by hepatocyte nuclear factor 4α (HNF4α). The HDAC3-PROX1 module controls the expression of a gene program regulating lipid homeostasis, and hepatic-specific ablation of either component increases triglyceride content in liver. These findings underscore the importance of specific combinations of transcription factors and coregulators in the fine tuning of organismal metabolism.HDAC3 is a critical mediator of hepatic lipid metabolism and its loss leads to fatty liver. Here, the authors characterize the liver HDAC3 interactome in vivo, provide evidence that HDAC3 interacts with PROX1, and show that HDAC3 and PROX1 control expression of genes regulating lipid homeostasis.


Assuntos
Fator 4 Nuclear de Hepatócito/metabolismo , Histona Desacetilases/metabolismo , Proteínas de Homeodomínio/metabolismo , Fígado/metabolismo , Triglicerídeos/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Regulação da Expressão Gênica , Fator 4 Nuclear de Hepatócito/genética , Histona Desacetilases/genética , Proteínas de Homeodomínio/genética , Lipídeos/genética , Masculino , Camundongos Knockout , Mapeamento de Interação de Proteínas/métodos , Proteínas Supressoras de Tumor/genética
5.
Science ; 348(6242): 1488-92, 2015 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-26044300

RESUMO

Circadian and metabolic physiology are intricately intertwined, as illustrated by Rev-erbα, a transcription factor (TF) that functions both as a core repressive component of the cell-autonomous clock and as a regulator of metabolic genes. Here, we show that Rev-erbα modulates the clock and metabolism by different genomic mechanisms. Clock control requires Rev-erbα to bind directly to the genome at its cognate sites, where it competes with activating ROR TFs. By contrast, Rev-erbα regulates metabolic genes primarily by recruiting the HDAC3 co-repressor to sites to which it is tethered by cell type-specific transcription factors. Thus, direct competition between Rev-erbα and ROR TFs provides a universal mechanism for self-sustained control of the molecular clock across all tissues, whereas Rev-erbα uses lineage-determining factors to convey a tissue-specific epigenomic rhythm that regulates metabolism tailored to the specific need of that tissue.


Assuntos
Proteínas CLOCK/genética , Relógios Circadianos/genética , Ritmo Circadiano/genética , Regulação da Expressão Gênica , Histona Desacetilases/metabolismo , Metabolismo/genética , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/metabolismo , Animais , Fator 6 Nuclear de Hepatócito/metabolismo , Metabolismo dos Lipídeos/genética , Fígado/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/genética , Membro 1 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Especificidade de Órgãos , Ligação Proteica , Distribuição Tecidual
6.
Cell ; 159(5): 1140-1152, 2014 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-25416951

RESUMO

Mammalian transcriptomes display complex circadian rhythms with multiple phases of gene expression that cannot be accounted for by current models of the molecular clock. We have determined the underlying mechanisms by measuring nascent RNA transcription around the clock in mouse liver. Unbiased examination of enhancer RNAs (eRNAs) that cluster in specific circadian phases identified functional enhancers driven by distinct transcription factors (TFs). We further identify on a global scale the components of the TF cistromes that function to orchestrate circadian gene expression. Integrated genomic analyses also revealed mechanisms by which a single circadian factor controls opposing transcriptional phases. These findings shed light on the diversity and specificity of TF function in the generation of multiple phases of circadian gene transcription in a mammalian organ.


Assuntos
Ritmo Circadiano , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica , Transcrição Gênica , Animais , Fatores de Transcrição de Zíper de Leucina Básica/genética , Relógios Circadianos , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/genética
7.
Cell Cycle ; 12(14): 2233-40, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23892437

RESUMO

SIRT1 is an NAD (+) -dependent deacetylase that counteracts multiple disease states associated with aging and may underlie some of the health benefits of calorie restriction. Understanding how SIRT1 is regulated in vivo could therefore lead to new strategies to treat age-related diseases. SIRT1 forms a stable complex with DBC1, an endogenous inhibitor. Little is known regarding the biochemical nature of SIRT1-DBC1 complex formation, how it is regulated and whether or not it is possible to block this interaction pharmacologically. In this study, we show that critical residues within the catalytic core of SIRT1 mediate binding to DBC1 via its N-terminal region, and that several carboxamide SIRT1 inhibitors, including EX-527, can completely block this interaction. We identify two acetylation sites on DBC1 that regulate its ability to bind SIRT1 and suppress its activity. Furthermore, we show that DBC1 itself is a substrate for SIRT1. Surprisingly, the effect of EX-527 on SIRT1-DBC1 binding is independent of DBC1 acetylation. Together, these data show that protein acetylation serves as an endogenous regulatory mechanism for SIRT1-DBC1 binding and illuminate a new path to developing small-molecule modulators of SIRT1.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Carbazóis/farmacologia , Regulação da Expressão Gênica , Inibidores de Histona Desacetilases/farmacologia , Processamento de Proteína Pós-Traducional , Sirtuína 1/metabolismo , Acetilação , Proteínas Adaptadoras de Transdução de Sinal/genética , Sítios de Ligação , Linhagem Celular Tumoral , Genes Reporter , Humanos , Luciferases/genética , Ligação Proteica/efeitos dos fármacos , Domínios e Motivos de Interação entre Proteínas , Transdução de Sinais , Sirtuína 1/antagonistas & inibidores , Sirtuína 1/genética , Especificidade por Substrato
8.
Science ; 339(6124): 1216-9, 2013 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-23471411

RESUMO

A molecule that treats multiple age-related diseases would have a major impact on global health and economics. The SIRT1 deacetylase has drawn attention in this regard as a target for drug design. Yet controversy exists around the mechanism of sirtuin-activating compounds (STACs). We found that specific hydrophobic motifs found in SIRT1 substrates such as PGC-1α and FOXO3a facilitate SIRT1 activation by STACs. A single amino acid in SIRT1, Glu(230), located in a structured N-terminal domain, was critical for activation by all previously reported STAC scaffolds and a new class of chemically distinct activators. In primary cells reconstituted with activation-defective SIRT1, the metabolic effects of STACs were blocked. Thus, SIRT1 can be directly activated through an allosteric mechanism common to chemically diverse STACs.


Assuntos
Sirtuína 1/química , Sirtuína 1/metabolismo , Estilbenos/farmacologia , Regulação Alostérica , Motivos de Aminoácidos , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Células Cultivadas , Ativação Enzimática , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/química , Fatores de Transcrição Forkhead/genética , Ácido Glutâmico/química , Ácido Glutâmico/genética , Compostos Heterocíclicos de 4 ou mais Anéis/química , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Interações Hidrofóbicas e Hidrofílicas , Camundongos , Dados de Sequência Molecular , Mioblastos/efeitos dos fármacos , Mioblastos/enzimologia , Estrutura Terciária de Proteína , Resveratrol , Sirtuína 1/genética , Estilbenos/química , Especificidade por Substrato
9.
Mol Cell Biol ; 33(8): 1487-502, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23382074

RESUMO

Although many functions and targets have been attributed to the histone and protein deacetylase SIRT1, a comprehensive analysis of SIRT1 binding proteins yielding a high-confidence interaction map has not been established. Using a comparative statistical analysis of binding partners, we have assembled a high-confidence SIRT1 interactome. Employing this method, we identified the deubiquitinating enzyme ubiquitin-specific protease 22 (USP22), a component of the deubiquitinating module (DUBm) of the SAGA transcriptional coactivating complex, as a SIRT1-interacting partner. We found that this interaction is highly specific, requires the ZnF-UBP domain of USP22, and is disrupted by the inactivating H363Y mutation within SIRT1. Moreover, we show that USP22 is acetylated on multiple lysine residues and that alteration of a single lysine (K129) within the ZnF-UBP domain is sufficient to alter interaction of the DUBm with the core SAGA complex. Furthermore, USP22-mediated recruitment of SIRT1 activity promotes the deacetylation of individual SAGA complex components. Our results indicate an important role of SIRT1-mediated deacetylation in regulating the formation of DUBm subcomplexes within the larger SAGA complex.


Assuntos
Mapas de Interação de Proteínas , Sirtuína 1/metabolismo , Tioléster Hidrolases/metabolismo , Acetilação , Linhagem Celular , Células HEK293 , Humanos , Mutação , Ligação Proteica , Processamento de Proteína Pós-Traducional , Interferência de RNA , RNA Interferente Pequeno , Sirtuína 1/genética , Tioléster Hidrolases/genética , Transativadores/genética , Transativadores/metabolismo , Ubiquitina Tiolesterase , Ubiquitinação
10.
Proc Natl Acad Sci U S A ; 108(43): E952-61, 2011 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-21949390

RESUMO

The formation of myelin by Schwann cells (SCs) occurs via a series of orchestrated molecular events. We previously used global expression profiling to examine peripheral nerve myelination and identified the NAD(+)-dependent deacetylase Sir-two-homolog 2 (Sirt2) as a protein likely to be involved in myelination. Here, we show that Sirt2 expression in SCs is correlated with that of structural myelin components during both developmental myelination and remyelination after nerve injury. Transgenic mice lacking or overexpressing Sirt2 specifically in SCs show delays in myelin formation. In SCs, we found that Sirt2 deacetylates Par-3, a master regulator of cell polarity. The deacetylation of Par-3 by Sirt2 decreases the activity of the polarity complex signaling component aPKC, thereby regulating myelin formation. These results demonstrate that Sirt2 controls an essential polarity pathway in SCs during myelin assembly and provide insights into the association between intracellular metabolism and SC plasticity.


Assuntos
Moléculas de Adesão Celular/metabolismo , Bainha de Mielina/fisiologia , Proteína Quinase C/metabolismo , Células de Schwann/fisiologia , Transdução de Sinais/fisiologia , Sirtuína 2/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Western Blotting , Proteínas de Ciclo Celular , Cromatografia Líquida , Primers do DNA/genética , Genótipo , Imunoprecipitação , Luciferases , Camundongos , Camundongos Transgênicos , Plasmídeos/genética , Reação em Cadeia da Polimerase em Tempo Real , Sirtuína 2/genética , Espectrometria de Massas em Tandem
11.
J Biol Chem ; 286(22): 19270-9, 2011 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-21467030

RESUMO

In mammals, the transcriptional activity of signal transducer and activator of transcription 3 (STAT3) is regulated by the deacetylase SIRT1. However, whether the newly described nongenomic actions of STAT3 toward mitochondrial oxidative phosphorylation are dependent on SIRT1 is unclear. In this study, Sirt1 gene knock-out murine embryonic fibroblast (MEF) cells were used to delineate the role of SIRT1 in the regulation of STAT3 mitochondrial function. Here, we show that STAT3 mRNA and protein levels and the accumulation of serine-phosphorylated STAT3 in mitochondria were increased significantly in Sirt1-KO cells as compared with wild-type MEFs. Various mitochondrial bioenergetic parameters, such as the oxygen consumption rate in cell cultures, enzyme activities of the electron transport chain complexes in isolated mitochondria, and production of ATP and lactate, indicated that Sirt1-KO cells exhibited higher mitochondrial respiration as compared with wild-type MEFs. Two independent approaches, including ectopic expression of SIRT1 and siRNA-mediated knockdown of STAT3, led to reduction in intracellular ATP levels and increased lactate production in Sirt1-KO cells that were approaching those of wild-type controls. Comparison of profiles of phospho-antibody array data indicated that the deletion of SirT1 was accompanied by constitutive activation of the pro-inflammatory NF-κB pathway, which is key for STAT3 induction and increased cellular respiration in Sirt1-KO cells. Thus, SIRT1 appears to be a functional regulator of NF-κB-dependent STAT3 expression that induces mitochondrial biogenesis. These results have implications for understanding the interplay between STAT3 and SIRT1 in pro-inflammatory conditions.


Assuntos
Embrião de Mamíferos/metabolismo , Fibroblastos/metabolismo , Mitocôndrias/metabolismo , Consumo de Oxigênio/fisiologia , Fator de Transcrição STAT3/biossíntese , Sirtuína 1/metabolismo , Trifosfato de Adenosina/biossíntese , Trifosfato de Adenosina/genética , Animais , Embrião de Mamíferos/citologia , Fibroblastos/citologia , Regulação da Expressão Gênica/fisiologia , Técnicas de Silenciamento de Genes , Ácido Láctico/metabolismo , Camundongos , Mitocôndrias/genética , NF-kappa B/genética , NF-kappa B/metabolismo , Fosforilação Oxidativa , Fosforilação , Fator de Transcrição STAT3/genética , Sirtuína 1/genética
12.
Aging (Albany NY) ; 1(6): 515-28, 2009 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-20157535

RESUMO

Resveratrol is a plant-derived polyphenol that promotes health and disease resistance in rodent models, and extends lifespan in lower organisms. A major challenge is to understand the biological processes and molecular pathways by which resveratrol induces these beneficial effects. Autophagy is a critical process by which cells turn over damaged components and maintain bioenergetic requirements. Disruption of the normal balance between pro- and anti-autophagic signals is linked to cancer, liver disease, and neurodegenerative disorders. Here we show that resveratrol attenuates autophagy in response to nutrient limitation or rapamycin in multiple cell lines through a pathway independent of a known target, SIRT1. In a large-scalein vitro kinase screen we identified p70 S6 kinase (S6K1) as a target of resveratrol. Blocking S6K1 activity by expression of a dominant-negative mutant or RNA interference is sufficient to disrupt autophagy to a similar extent as resveratrol. Furthermore, co-administration of resveratrol with S6K1 knockdown does not produce an additive effect. These data indicate that S6K1 is important for the full induction of autophagy in mammals and raise the possibility that some of the beneficial effects of resveratrol are due to modulation of S6K1 activity.


Assuntos
Autofagia/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas 70-kDa/antagonistas & inibidores , Estilbenos/farmacologia , Animais , Autofagia/fisiologia , Linhagem Celular , Humanos , Camundongos , Quercetina , Resveratrol , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Sirolimo/farmacologia , Estilbenos/química
13.
EMBO J ; 26(13): 3169-79, 2007 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-17581637

RESUMO

A progressive loss of neurons with age underlies a variety of debilitating neurological disorders, including Alzheimer's disease (AD) and amyotrophic lateral sclerosis (ALS), yet few effective treatments are currently available. The SIR2 gene promotes longevity in a variety of organisms and may underlie the health benefits of caloric restriction, a diet that delays aging and neurodegeneration in mammals. Here, we report that a human homologue of SIR2, SIRT1, is upregulated in mouse models for AD, ALS and in primary neurons challenged with neurotoxic insults. In cell-based models for AD/tauopathies and ALS, SIRT1 and resveratrol, a SIRT1-activating molecule, both promote neuronal survival. In the inducible p25 transgenic mouse, a model of AD and tauopathies, resveratrol reduced neurodegeneration in the hippocampus, prevented learning impairment, and decreased the acetylation of the known SIRT1 substrates PGC-1alpha and p53. Furthermore, injection of SIRT1 lentivirus in the hippocampus of p25 transgenic mice conferred significant protection against neurodegeneration. Thus, SIRT1 constitutes a unique molecular link between aging and human neurodegenerative disorders and provides a promising avenue for therapeutic intervention.


Assuntos
Doença de Alzheimer/metabolismo , Esclerose Lateral Amiotrófica/metabolismo , Degeneração Neural/metabolismo , Sirtuínas/metabolismo , Acetilação/efeitos dos fármacos , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/patologia , Animais , Células Cultivadas , Quinase 5 Dependente de Ciclina/metabolismo , Modelos Animais de Doenças , Ativação Enzimática , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Transgênicos , Mutação/genética , Degeneração Neural/genética , Degeneração Neural/patologia , Ratos , Resveratrol , Sirtuína 1 , Sirtuínas/genética , Estilbenos/farmacologia , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1 , Proteína Supressora de Tumor p53/metabolismo
14.
Cancer Cell ; 7(1): 77-85, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15652751

RESUMO

Several mitochondrial proteins are tumor suppressors. These include succinate dehydrogenase (SDH) and fumarate hydratase, both enzymes of the tricarboxylic acid (TCA) cycle. However, to date, the mechanisms by which defects in the TCA cycle contribute to tumor formation have not been elucidated. Here we describe a mitochondrion-to-cytosol signaling pathway that links mitochondrial dysfunction to oncogenic events: succinate, which accumulates as a result of SDH inhibition, inhibits HIF-alpha prolyl hydroxylases in the cytosol, leading to stabilization and activation of HIF-1alpha. These results suggest a mechanistic link between SDH mutations and HIF-1alpha induction, providing an explanation for the highly vascular tumors that develop in the absence of VHL mutations.


Assuntos
Ciclo do Ácido Cítrico/fisiologia , Oncogenes , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Ácido Succínico/metabolismo , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular , Transformação Celular Neoplásica , Ativação Enzimática , Regulação Enzimológica da Expressão Gênica , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia , Mitocôndrias/metabolismo , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Pró-Colágeno-Prolina Dioxigenase/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/fisiologia , Succinato Desidrogenase/antagonistas & inibidores , Succinato Desidrogenase/genética , Succinato Desidrogenase/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Proteína Supressora de Tumor Von Hippel-Lindau
15.
Proc Natl Acad Sci U S A ; 99(20): 12801-6, 2002 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-12228733

RESUMO

The ability of cells to maintain a bioenergetically favorable ATP/ADP ratio confers a tight balance between cellular events that consume ATP and the rate of ATP production. However, after growth factor withdrawal, the cellular ATP/ADP ratio declines. To investigate these changes, mitochondria from growth factor-deprived cells isolated before the onset of apoptosis were characterized in vitro. Mitochondria from growth factor-deprived cells have lost their ability to undergo matrix condensation in response to ADP, which is accompanied by a failure to perform ADP-coupled respiration. At the time of analysis, mitochondria from growth factor-deprived cells were not depleted of cytochrome c and cytochrome c-dependent respiration was unaffected, demonstrating that the inhibition of the respiratory rate is not due to loss of cytochrome c. Agents that disrupt the mitochondrial outer membrane, such as digitonin, or maintain outer membrane exchange of adenine nucleotide, such as Bcl-x(L), restored ADP-dependent control of mitochondrial respiration. Together, these data suggest that the regulation of mitochondrial outer membrane permeability contributes to respiratory control.


Assuntos
Substâncias de Crescimento/fisiologia , Interleucina-3/fisiologia , Mitocôndrias/metabolismo , Consumo de Oxigênio , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Apoptose , Linhagem Celular , Membrana Celular/metabolismo , Meios de Cultura Livres de Soro/farmacologia , Grupo dos Citocromos c/metabolismo , Interleucina-3/metabolismo , Camundongos , Microscopia Eletrônica , Oxigênio/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína bcl-X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...