Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Cell Endocrinol ; 408: 62-72, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25724481

RESUMO

High output of steroid hormone synthesis in steroidogenic cells of the adrenal cortex and the gonads requires the expression of the steroidogenic acute regulatory protein (StAR) that facilitates cholesterol mobilization to the mitochondrial inner membrane where the CYP11A1/P450scc enzyme complex converts the sterol to the first steroid. Earlier studies have shown that StAR is active while pausing on the cytosolic face of the outer mitochondrial membrane while subsequent import of the protein into the matrix terminates the cholesterol mobilization activity. Consequently, during repeated activity cycles, high level of post-active StAR accumulates in the mitochondrial matrix. To prevent functional damage due to such protein overload effect, StAR is degraded by a sequence of three to four ATP-dependent proteases of the mitochondria protein quality control system, including LON and the m-AAA membranous proteases AFG3L2 and SPG7/paraplegin. Furthermore, StAR expression in both peri-ovulatory ovarian cells, or under ectopic expression in cell line models, results in up to 3-fold enrichment of the mitochondrial proteases and their transcripts. We named this novel form of mitochondrial stress as StAR overload response (SOR). To better understand the SOR mechanism at the transcriptional level we analyzed first the unexplored properties of the proximal promoter of the LON gene. Our findings suggest that the human nuclear respiratory factor 2 (NRF-2), also known as GA binding protein (GABP), is responsible for 88% of the proximal promoter activity, including the observed increase of transcription in the presence of StAR. Further studies are expected to reveal if common transcriptional determinants coordinate the SOR induced transcription of all the genes encoding the SOR proteases.


Assuntos
Fator de Transcrição de Proteínas de Ligação GA/metabolismo , Mitocôndrias/metabolismo , Fosfoproteínas/metabolismo , Protease La/genética , Estresse Fisiológico/genética , Ativação Transcricional/genética , Animais , Sequência de Bases , Humanos , Dados de Sequência Molecular
2.
Mol Endocrinol ; 28(2): 208-24, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24422629

RESUMO

Steroidogenic acute regulatory protein (StAR) is essential for steroid hormone synthesis in the adrenal cortex and the gonads. StAR activity facilitates the supply of cholesterol substrate into the inner mitochondrial membranes where conversion of the sterol to a steroid is catalyzed. Mitochondrial import terminates the cholesterol mobilization activity of StAR and leads to mounting accumulation of StAR in the mitochondrial matrix. Our studies suggest that to prevent mitochondrial impairment, StAR proteolysis is executed by at least 2 mitochondrial proteases, ie, the matrix LON protease and the inner membrane complexes of the metalloproteases AFG3L2 and AFG3L2:SPG7/paraplegin. Gonadotropin administration to prepubertal rats stimulated ovarian follicular development associated with increased expression of the mitochondrial protein quality control system. In addition, enrichment of LON and AFG3L2 is evident in StAR-expressing ovarian cells examined by confocal microscopy. Furthermore, reporter studies of the protease promoters examined in the heterologous cell model suggest that StAR expression stimulates up to a 3.5-fold increase in the protease gene transcription. Such effects are StAR-specific, are independent of StAR activity, and failed to occur upon expression of StAR mutants that do not enter the matrix. Taken together, the results of this study suggest the presence of a novel regulatory loop, whereby acute accumulation of an apparent nuisance protein in the matrix provokes a mitochondria to nucleus signaling that, in turn, activates selected transcription of genes encoding the enrichment of mitochondrial proteases relevant for enhanced clearance of StAR.


Assuntos
Proteases Dependentes de ATP/genética , Metaloendopeptidases/genética , Mitocôndrias/enzimologia , Fosfoproteínas/fisiologia , Proteases Dependentes de ATP/metabolismo , ATPases Associadas a Diversas Atividades Celulares , Animais , Células COS , Chlorocebus aethiops , Indução Enzimática , Feminino , Células HEK293 , Células HeLa , Humanos , Metaloendopeptidases/metabolismo , Ovário/enzimologia , Regiões Promotoras Genéticas , Protease La/genética , Protease La/metabolismo , Proteólise , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transcrição Gênica
3.
J Mol Neurosci ; 53(3): 306-15, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24258317

RESUMO

Selective serotonin reuptake inhibitors (SSRIs) show anti-inflammatory effects, suggesting a possible interaction with both Toll-like-receptor 4 (TLR4) responses and cholinergic signaling through as yet unclear molecular mechanism(s). Our results of structural modeling support the concept that the antidepressant fluoxetine physically interacts with the TLR4-myeloid differentiation factor-2 complex at the same site as bacterial lipopolysaccharide (LPS). We also demonstrate reduced LPS-induced pro-inflammatory interleukin-6 and tumor necrosis factor alpha in human peripheral blood mononuclear cells preincubated with fluoxetine. Furthermore, we show that fluoxetine intercepts the LPS-induced decreases in intracellular acetylcholinesterase (AChE-S) and that AChE-S interacts with the nuclear factor kappa B (NFκB)-activating intracellular receptor for activated C kinase 1 (RACK1). This interaction may prevent NFκB activation by residual RACK1 and its interacting protein kinase PKCßII. Our findings attribute the anti-inflammatory properties of SSRI to surface membrane interference with leukocyte TLR4 activation accompanied by intracellular limitation of pathogen-inducible changes in AChE-S, RACK1, and PKCßII.


Assuntos
Acetilcolinesterase/metabolismo , Anti-Inflamatórios/farmacologia , Fluoxetina/farmacologia , Proteínas de Ligação ao GTP/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Superfície Celular/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Acetilcolinesterase/química , Sequência de Aminoácidos , Sítios de Ligação , Proteínas Ligadas por GPI/química , Proteínas Ligadas por GPI/metabolismo , Proteínas de Ligação ao GTP/química , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Lipopolissacarídeos/farmacologia , Simulação de Acoplamento Molecular , Dados de Sequência Molecular , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Proteínas de Neoplasias/química , Ligação Proteica , Proteína Quinase C beta/metabolismo , Receptores de Quinase C Ativada , Receptores de Superfície Celular/química , Receptor 4 Toll-Like/química , Receptor 4 Toll-Like/metabolismo
4.
Mol Endocrinol ; 22(4): 951-64, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18187601

RESUMO

The steroidogenic acute regulatory protein (StAR) stimulates the regulated production of steroid hormones in the adrenal cortex and gonads by facilitating the delivery of cholesterol to the inner mitochondrial membrane. To explore key aspects of StAR function within bona fide steroidogenic cells, we used a transgenic mouse model to explore the function of StAR proteins in vivo. We first validated this transgenic bacterial artificial chromosome reconstitution system by targeting enhanced green fluorescent protein to steroidogenic cells of the adrenal cortex and gonads. Thereafter, we targeted expression of either wild-type StAR (WT-StAR) or a mutated StAR protein lacking the mitochondrial targeting signal (N47-StAR). In the context of mice homozygous for a StAR knockout allele (StAR-/-), all StAR activity derived from the StAR transgenes, allowing us to examine the function of the proteins that they encode. The WT-StAR transgene consistently restored viability and steroidogenic function to StAR-/- mice. Although the N47-StAR protein was reportedly active in transfected COS cells and mitochondrial reconstitution experiments, the N47-StAR transgene rescued viability in only 40% of StAR-/- mice. Analysis of lipid deposits in the primary steroidogenic tissues revealed a hierarchy of StAR function provided by N47-StAR: florid lipid deposits were seen in the adrenal cortex and ovarian theca region, with milder deposits in the Leydig cells. Our results confirm the ability of StAR lacking its mitochondrial targeting signal to perform some essential functions in vivo but also demonstrate important functional defects that differ from in vitro studies obtained in nonsteroidogenic cells.


Assuntos
Cromossomos Artificiais Bacterianos/genética , Mitocôndrias/metabolismo , Fosfoproteínas/fisiologia , Glândulas Suprarrenais/metabolismo , Hormônio Adrenocorticotrópico/sangue , Animais , Southern Blotting , Corticosterona/sangue , Feminino , Técnicas de Transferência de Genes , Gônadas/metabolismo , Immunoblotting , Masculino , Camundongos , Camundongos Transgênicos , Modelos Genéticos , Ovário/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Transporte Proteico/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Testículo/metabolismo
5.
Mol Endocrinol ; 21(9): 2164-77, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17579211

RESUMO

Steroidogenic acute regulatory protein (StAR) is a vital mitochondrial protein promoting transfer of cholesterol into steroid making mitochondria in specialized cells of the adrenal cortex and gonads. Our previous work has demonstrated that StAR is rapidly degraded upon import into the mitochondrial matrix. To identify the protease(s) responsible for this rapid turnover, murine StAR was expressed in wild-type Escherichia coli or in mutant strains lacking one of the four ATP-dependent proteolytic systems, three of which are conserved in mammalian mitochondria-ClpP, FtsH, and Lon. StAR was rapidly degraded in wild-type bacteria and stabilized only in lon (-)mutants; in such cells, StAR turnover was fully restored upon coexpression of human mitochondrial Lon. In mammalian cells, the rate of StAR turnover was proportional to the cell content of Lon protease after expression of a Lon-targeted small interfering RNA, or overexpression of the protein. In vitro assays using purified proteins showed that Lon-mediated degradation of StAR was ATP-dependent and blocked by the proteasome inhibitors MG132 (IC(50) = 20 microm) and clasto-lactacystin beta-lactone (cLbetaL, IC(50) = 3 microm); by contrast, epoxomicin, representing a different class of proteasome inhibitors, had no effect. Such inhibition is consistent with results in cultured rat ovarian granulosa cells demonstrating that degradation of StAR in the mitochondrial matrix is blocked by MG132 and cLbetaL but not by epoxomicin. Both inhibitors also blocked Lon-mediated cleavage of the model substrate fluorescein isothiocyanate-casein. Taken together, our former studies and the present results suggest that Lon is the primary ATP-dependent protease responsible for StAR turnover in mitochondria of steroidogenic cells.


Assuntos
Mitocôndrias/metabolismo , Fosfoproteínas/metabolismo , Protease La/fisiologia , Inibidores de Proteassoma , Trifosfato de Adenosina/fisiologia , Animais , Células Cultivadas , Feminino , Hormônios Esteroides Gonadais/biossíntese , Células da Granulosa/metabolismo , Camundongos , Fosfoproteínas/genética , Ratos , Ratos Sprague-Dawley
6.
Mol Cell Endocrinol ; 265-266: 51-8, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17218054

RESUMO

Steroidogenic acute regulatory protein (StAR) is a mitochondrial protein essential for massive synthesis of steroid hormones in the adrenal and the gonads. Our studies suggest that once synthesized on free polyribosomes, StAR preprotein either associates with the outer mitochondrial membrane to mediate transfer of cholesterol substrate required for steroidgenesis, or it is degraded by the proteasome. Proteasome inhibitors can prevent the turnover of StAR preprotein and other matrix-targeted preproteins. Once imported, excessive accumulation of inactive StAR in the matrix is avoided by a rapid turnover. Unexpectedly, mitochondrial StAR turnover can be inhibited by two proteasome inhibitors, i.e., MG132 and clasto-lactacystin beta-lactone, but not epoxomicin. Use of those inhibitors and immuno-electron microscopy data enabled a clear distinction between two pools of intra-mitochondrial StAR, one degraded by matrix protease(s) shortly after import, while the rest of the protein undergoes a slower and inhibitor resistant degradation following translocation onto to the matrix face of the inner membranes.


Assuntos
Mitocôndrias/enzimologia , Peptídeo Hidrolases/metabolismo , Fosfoproteínas/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Animais , Humanos , Lactonas/farmacologia , Leupeptinas/farmacologia , Mitocôndrias/metabolismo , Membranas Mitocondriais/enzimologia , Membranas Mitocondriais/metabolismo , Oligopeptídeos/farmacologia , Inibidores de Proteases/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...