Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
iScience ; 26(8): 107375, 2023 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-37599829

RESUMO

Spinal and bulbar muscular atrophy (SBMA) is a neuromuscular disease with substantial mitochondrial and metabolic dysfunctions. SBMA is caused by polyglutamine (polyQ) expansion in the androgen receptor (AR). Activating or increasing the NAD+-dependent deacetylase, SIRT3, reduced oxidative stress and death of cells modeling SBMA. However, increasing diminished SIRT3 in AR100Q mice failed to reduce acetylation of the SIRT3 target/antioxidant, SOD2, and had no effect on increased total acetylated peptides in quadriceps. Yet, overexpressing SIRT3 resulted in a trend of motor recovery, and corrected TCA cycle activity by decreasing acetylation of SIRT3 target proteins. We sought to boost blunted SIRT3 activity by replenishing diminished NAD+ with PARP inhibition. Although NAD+ was not affected, overexpressing SIRT3 with PARP inhibition fully restored hexokinase activity, correcting the glycolytic pathway in AR100Q quadriceps, and rescued motor endurance of SBMA mice. These data demonstrate that targeting metabolic anomalies can restore motor function downstream of polyQ-expanded AR.

2.
Front Mol Neurosci ; 10: 159, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28596723

RESUMO

Spinal and bulbar muscular atrophy (SBMA) is caused by expression of a polyglutamine (polyQ)-expanded androgen receptor (AR). The inefficient nuclear proteasomal degradation of the mutant AR results in the formation of nuclear inclusions containing amino-terminal fragments of the mutant AR. PA28γ (also referred to as REGγ) is a nuclear 11S-proteasomal activator with limited proteasome activation capabilities compared to its cytoplasmic 11S (PA28α, PA28ß) counterparts. To clarify the role of REGγ in polyQ-expanded AR metabolism, we carried out genetic and biochemical studies in cell models of SBMA. Overexpression of REGγ in a PC12 cell model of SBMA increased polyQ-expanded AR aggregation and contributed to polyQ-expanded AR toxicity in the presence of dihydrotestosterone (DHT). These effects of REGγ were independent of its association with the proteasome and may be due, in part, to the decreased binding of polyQ-expanded AR by the E3 ubiquitin-ligase MDM2. Unlike its effects in PC12 cells, REGγ overexpression rescued transgenic SBMA motor neurons from DHT-induced toxicity in a proteasome binding-dependent manner, suggesting that the degradation of a specific 11S proteasome substrate or substrates promotes motor neuron viability. One potential substrate that we found to play a role in mutant AR toxicity is the splicing factor SC35. These studies reveal that, depending on the cellular context, two biological roles for REGγ impact cell viability in the face of polyQ-expanded AR; a proteasome binding-independent mechanism directly promotes mutant AR aggregation while a proteasome binding-dependent mechanism promotes cell viability. The balance between these functions likely determines REGγ effects on polyQ-expanded AR-expressing cells.

3.
Brain Res ; 1628(Pt B): 254-264, 2015 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-26453288

RESUMO

Polyglutamine-repeat disorders are part of a larger family of neurodegenerative diseases characterized by protein misfolding and aggregation. In spinal and bulbar muscular atrophy (SBMA), polyglutamine expansion within the androgen receptor (AR) causes progressive debilitating muscular atrophy and lower motor neuron loss in males. Although soluble polyglutamine-expanded aggregation species are considered toxic intermediates in the aggregation process, relatively little is known about the spectrum of structures that are formed. Here we identify novel polyglutamine-expanded AR aggregates that are SDS-soluble and bind the toxicity-predicting antibody 3B5H10. Soluble, 3B5H10-reactive aggregation species exist in low-density conformations and are larger by atomic force microscopy, suggesting that they may be less compact than later-stage, insoluble aggregates. We demonstrate disease-relevance in vivo and draw correlations with toxicity in vitro. This article is part of a Special Issue entitled SI: Neuroprotection.


Assuntos
Transtornos Musculares Atróficos/genética , Transtornos Musculares Atróficos/patologia , Peptídeos/genética , Peptídeos/metabolismo , Receptores Androgênicos/genética , Medula Espinal/metabolismo , Animais , Anticorpos/metabolismo , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Modelos Animais de Doenças , Humanos , Imunoprecipitação , Masculino , Camundongos , Camundongos Transgênicos , Microscopia de Força Atômica , Células PC12 , Peptídeos/imunologia , Ratos , Transfecção
4.
J Clin Invest ; 125(2): 831-45, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25607844

RESUMO

Expansion of the polyglutamine (polyQ) tract within the androgen receptor (AR) causes neuromuscular degeneration in individuals with spinobulbar muscular atrophy (SBMA). PolyQ AR has diminished transcriptional function and exhibits ligand-dependent proteotoxicity, features that have both been implicated in SBMA; however, the extent to which altered AR transcriptional function contributes to pathogenesis remains controversial. Here, we sought to dissociate effects of diminished AR function from polyQ-mediated proteotoxicity by enhancing the transcriptional activity of polyQ AR. To accomplish this, we bypassed the inhibitory effect of AR SUMOylation (where SUMO indicates small ubiquitin-like modifier) by mutating conserved lysines in the polyQ AR that are sites of SUMOylation. We determined that replacement of these residues by arginine enhances polyQ AR activity as a hormone-dependent transcriptional regulator. In a murine model, disruption of polyQ AR SUMOylation rescued exercise endurance and type I muscle fiber atrophy; it also prolonged survival. These changes occurred without overt alterations in polyQ AR expression or aggregation, revealing the favorable trophic support exerted by the ligand-activated receptor. Our findings demonstrate beneficial effects of enhancing the transcriptional function of the ligand-activated polyQ AR and indicate that the SUMOylation pathway may be a potential target for therapeutic intervention in SBMA.


Assuntos
Fibras Musculares de Contração Lenta/metabolismo , Transtornos Musculares Atróficos/metabolismo , Peptídeos/metabolismo , Receptores Androgênicos/metabolismo , Sumoilação , Transcrição Gênica , Animais , Camundongos , Camundongos Transgênicos , Fibras Musculares de Contração Lenta/patologia , Transtornos Musculares Atróficos/genética , Transtornos Musculares Atróficos/patologia , Células PC12 , Peptídeos/genética , Ratos , Receptores Androgênicos/genética
6.
J Neurosci ; 31(48): 17425-36, 2011 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-22131404

RESUMO

Posttranslational protein modifications can play a major role in disease pathogenesis; phosphorylation, sumoylation, and acetylation modulate the toxicity of a variety of proteotoxic proteins. The androgen receptor (AR) is substantially modified, in response to hormone binding, by phosphorylation, sumoylation, and acetylation; these modifications might thus contribute to DHT-dependent polyglutamine (polyQ)-expanded AR proteotoxicity in spinal and bulbar muscular atrophy (SBMA). SIRT1, a nuclear protein and deacetylase of the AR, is neuroprotective in many neurodegenerative disease models. Our studies reveal that SIRT1 also offers protection against polyQ-expanded AR by deacetylating the AR at lysines 630/632/633. This finding suggested that nuclear AR acetylation plays a role in the aberrant metabolism and toxicity of polyQ-expanded AR. Subsequent studies revealed that the polyQ-expanded AR is hyperacetylated and that pharmacologic reduction of acetylation reduces mutant AR aggregation. Moreover, genetic mutation to inhibit polyQ-expanded AR acetylation of lysines 630/632/633 substantially decreased its aggregation and completely abrogated its toxicity in cell lines and motor neurons. Our studies also reveal one means by which the AR acetylation state likely modifies polyQ-expanded AR metabolism and toxicity, through its effect on DHT-dependent AR stabilization. Overall, our findings reveal a neuroprotective function of SIRT1 that operates through its deacetylation of polyQ-expanded AR and highlight the potential of both SIRT1 and AR acetylation as powerful therapeutic targets in SBMA.


Assuntos
Atrofia Muscular Espinal/genética , Receptores Androgênicos/genética , Sirtuína 1/genética , Acetilação , Animais , Núcleo Celular/genética , Núcleo Celular/metabolismo , Modelos Animais de Doenças , Camundongos , Atrofia Muscular Espinal/metabolismo , Neurônios/metabolismo , Células PC12 , Transporte Proteico/genética , Ratos , Receptores Androgênicos/metabolismo , Sirtuína 1/metabolismo , Medula Espinal/metabolismo , Expansão das Repetições de Trinucleotídeos
7.
J Biol Chem ; 285(46): 35567-77, 2010 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-20826791

RESUMO

Polyglutamine expansion within the androgen receptor (AR) causes spinal and bulbar muscular atrophy (SBMA) and is associated with misfolded and aggregated species of the mutant AR. We showed previously that nuclear localization of the mutant AR was necessary but not sufficient for SBMA. Here we show that an interdomain interaction of the AR that is central to its function within the nucleus is required for AR aggregation and toxicity. Ligands that prevent the interaction between the amino-terminal FXXLF motif and carboxyl-terminal AF-2 domain (N/C interaction) prevented toxicity and AR aggregation in an SBMA cell model and rescued primary SBMA motor neurons from 5α-dihydrotestosterone-induced toxicity. Moreover, genetic mutation of the FXXLF motif prevented AR aggregation and 5α-dihydrotestosterone toxicity. Finally, selective androgen receptor modulators, which prevent the N/C interaction, ameliorated AR aggregation and toxicity while maintaining AR function, highlighting a novel therapeutic strategy to prevent the SBMA phenotype while retaining AR transcriptional function.


Assuntos
Mutação , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Expansão das Repetições de Trinucleotídeos/genética , Motivos de Aminoácidos/genética , Sequência de Aminoácidos , Antagonistas de Androgênios/farmacologia , Androgênios/farmacologia , Anilidas/farmacologia , Animais , Sítios de Ligação/genética , Western Blotting , Atrofia Bulboespinal Ligada ao X/genética , Atrofia Bulboespinal Ligada ao X/metabolismo , Atrofia Bulboespinal Ligada ao X/patologia , Células Cultivadas , Di-Hidrotestosterona/farmacologia , Células HEK293 , Humanos , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Neurônios Motores/citologia , Neurônios Motores/metabolismo , Nitrilas/farmacologia , Células PC12 , Ligação Proteica/efeitos dos fármacos , Ratos , Receptores Androgênicos/química , Testosterona/farmacologia , Compostos de Tosil/farmacologia , Técnicas do Sistema de Duplo-Híbrido
8.
Autophagy ; 5(8): 1194-7, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19770590

RESUMO

Ridding neurons of toxic misfolded proteins is a critical feature of many neurodegenerative diseases. We have recently reported that lack of access of nuclear polyglutamine-expanded androgen receptor (AR) to the autophagic degradation pathway is a critical point in pathogenesis. When mutant AR is contained within the cytoplasm, it can be degraded by autophagy, resulting in amelioration of its toxic effects, as has been observed in other polyglutamine expansion diseases involving cytoplasmic mutant proteins. However, we have also found that pharmacological induction of autophagy protects SBMA motor neurons from the toxic effects of even nuclear localized mutant AR, albeit without affecting mutant nuclear AR levels. Thus, we have further investigated the mechanism by which autophagy elicits therapeutic benefit in cell culture. We found that endogenous autophagy only slightly alters nuclear mutant AR aggregation compared to substantial effects on cytoplasmic AR aggregation. Interestingly, pharmacological activation of mTOR-dependent autophagy did not significantly alter nuclear AR aggregation, whereas we observed that it protects SBMA motor neurons. Our findings indicate that therapeutic intervention to induce autophagy represents a potential potent benefit for SBMA, and that it likely does so by protecting SBMA motor neurons independent of a direct effect on mutant AR.


Assuntos
Autofagia , Atrofia Bulboespinal Ligada ao X/metabolismo , Atrofia Bulboespinal Ligada ao X/patologia , Receptores Androgênicos/metabolismo , Animais , Autofagia/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Camundongos , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Células PC12 , Peptídeos/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Transporte Proteico/efeitos dos fármacos , Ratos , Frações Subcelulares/metabolismo , Trealose/farmacologia
9.
Hum Mol Genet ; 18(11): 1937-50, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19279159

RESUMO

The nucleus is the primary site of protein aggregation in many polyglutamine diseases, suggesting a central role in pathogenesis. In SBMA, the nucleus is further implicated by the critical role for disease of androgens, which promote the nuclear translocation of the mutant androgen receptor (AR). To clarify the importance of the nucleus in SBMA, we genetically manipulated the nuclear localization signal of the polyglutamine-expanded AR. Transgenic mice expressing this mutant AR displayed inefficient nuclear translocation and substantially improved motor function compared with SBMA mice. While we found that nuclear localization of polyglutamine-expanded AR is required for SBMA, we also discovered, using cell models of SBMA, that it is insufficient for both aggregation and toxicity and requires androgens for these disease features. Through our studies of cultured motor neurons, we further found that the autophagic pathway was able to degrade cytoplasmically retained expanded AR and represents an endogenous neuroprotective mechanism. Moreover, pharmacologic induction of autophagy rescued motor neurons from the toxic effects of even nuclear-residing mutant AR, suggesting a therapeutic role for autophagy in this nucleus-centric disease. Thus, our studies firmly establish that polyglutamine-expanded AR must reside within nuclei in the presence of its ligand to cause SBMA. They also highlight a mechanistic basis for the requirement for nuclear localization in SBMA neurotoxicity, namely the lack of mutant AR removal by the autophagic protein degradation pathway.


Assuntos
Autofagia , Citoplasma/metabolismo , Atrofia Muscular Espinal/fisiopatologia , Peptídeos/metabolismo , Receptores Androgênicos/metabolismo , Expansão das Repetições de Trinucleotídeos , Androgênios/metabolismo , Animais , Linhagem Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Células Cultivadas , Citoplasma/genética , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Atividade Motora , Neurônios Motores/citologia , Neurônios Motores/metabolismo , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/metabolismo , Sinais de Localização Nuclear/genética , Peptídeos/genética , Transporte Proteico , Receptores Androgênicos/genética
10.
Resuscitation ; 66(3): 379-89, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16029920

RESUMO

Visceral organs display differential sensitivity to ischemia and reperfusion injury, but the cellular mechanisms underlying these differential responses are not completely understood. A significant response to ischemia identified in brain is stress to the endoplasmic reticulum (ER), as indicated by PKR-like endoplasmic reticulum eIF2alpha kinase (PERK)-mediated phosphorylation of eIF2alpha. To determine the generality of this response, we evaluated the PERK pathway in brain, GI tract, heart, liver, lung, kidney, pancreas and skeletal muscle following a clinically relevant, 10 min cardiac arrest-induced whole body ischemia and either 10 or 90 min reperfusion. The potential role of nitric oxide (NO) on PERK activation was investigated by conducting ischemia and reperfusion in the presence and absence of the NO synthase inhibitor nitro-L-arginine methyl ester (L-NAME). Organ stress could be ranked with respect to the degree of eIF2alpha phosphorylation at 10 min reperfusion. Brain, kidney and GI tract were reactive organs, showing 15 to 20-fold increases in eIF2alpha(P) compared to controls. Moderately reactive organs included liver and heart, showing <10-fold increases in eIF2alpha(P). Pancreas, lung and skeletal muscle were nonreactive. Although treatment of cultured neuroblastoma 104 cells with the NO-donor S-nitroso-N-acetyl-penicillamine (SNAP) activated PERK, administration of L-NAME had no effect on PERK activation or eIF2alpha phosphorylation in organs following ischemia and reperfusion. Thus, PERK is activated differentially in reperfused organs independent of NO. These results suggest that ER stress may play a role in differential responses of viscera to ischemia and reperfusion. ER stress in viscera may contribute to the pathophysiology of resuscitation from cardiac arrest and during organ transplantation procedures.


Assuntos
Reanimação Cardiopulmonar , Parada Cardíaca/enzimologia , Parada Cardíaca/terapia , eIF-2 Quinase/metabolismo , Animais , Encéfalo/enzimologia , Modelos Animais de Doenças , Fator de Iniciação 2 em Eucariotos/metabolismo , Trato Gastrointestinal/enzimologia , Parada Cardíaca/complicações , Rim/enzimologia , Fígado/enzimologia , Pulmão/enzimologia , Masculino , Músculo Esquelético/enzimologia , Doadores de Óxido Nítrico/farmacologia , Pâncreas/enzimologia , Penicilamina/análogos & derivados , Penicilamina/farmacologia , Ratos , Ratos Long-Evans , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/etiologia , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/enzimologia
11.
Biochim Biophys Acta ; 1741(3): 314-24, 2005 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-15936177

RESUMO

Inhibition of protein synthesis occurs in the post-ischemic reperfused kidney but the molecular mechanism of renal translation arrest is unknown. Several pathways have been identified whereby cell stress inhibits translation initiation via phosphorylation of the alpha subunit of eukaryotic initiation factor 2 (eIF 2 alpha, phospho-form eIF 2 alpha(P)]. Here, we report a 20-fold increase in eIF 2 alpha(P) in kidney homogenates following 10 min of cardiac arrest-induced ischemia and 10 min reperfusion. Using immunohistochemistry, we observed eIF 2 alpha(P) in tubular epithelial cells in both cortex and medulla, where the greatest eIF 2 alpha(P) staining was found in epithelial cells of the so-called watershed area at the corticomedullary junction. We further show that increased eIF 2 alpha(P) is accompanied by activation of the PKR-like endoplasmic reticulum eIF 2 alpha kinase (PERK). These observations indicate that renal ischemia and reperfusion induce stress to the endoplasmic reticulum and activate the unfolded protein response in renal epithelial cells. As the unfolded protein response can result alternatively in a pro-survival or pro-apoptotic outcome, the present study demonstrates an new additional mechanism involved in cell damage and/or repair in ischemic and reperfused kidney.


Assuntos
Células Epiteliais/metabolismo , Regulação Enzimológica da Expressão Gênica , Rim/irrigação sanguínea , Traumatismo por Reperfusão/enzimologia , eIF-2 Quinase/metabolismo , Animais , Western Blotting , Creatina/sangue , Imuno-Histoquímica , Imunoprecipitação , Rim/metabolismo , Masculino , Ratos , Ratos Long-Evans
12.
J Neurochem ; 91(1): 1-8, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15379881

RESUMO

We review studies of endoplasmic reticulum (ER) stress and the unfolded protein response (UPR) following cerebral ischemia and reperfusion (I/R). The UPR is a cell stress program activated when misfolded proteins accumulate in the ER lumen. UPR activation causes: (i) a PERK-mediated phosphorylation of eIF2alpha, inhibiting protein synthesis to prevent further accumulation of unfolded proteins in the ER and (ii) upregulation of genes coding for ER-resident enzymes and chaperones and others, via eIF2alpha(p), and ATF6 and IRE1 activation. UPR-induced transcription increases capacity of the ER to process misfolded proteins. If ER stress and the UPR are prolonged, apoptosis ensues. Multiple forms of ER stress have been observed following brain I/R. The UPR following brain I/R is not isomorphic between in vivo I/R models and in vitro cell culture systems with pharmacological UPR induction. Although PERK and IRE1 are activated in the initial hours of reperfusion, total PERK decreases, ATF6 is not activated, and there is delayed appearance of UPR-induced mRNAs. Thus, multiple damage mechanisms associated with brain I/R alter UPR expression and contribute to a pro-apoptotic phenotype in neurons. Insights resulting from these studies will be important for the development of therapies to halt neuronal death following brain I/R.


Assuntos
Isquemia Encefálica/metabolismo , Chaperonas Moleculares/química , Dobramento de Proteína , Animais , Proteínas de Ligação a DNA/metabolismo , Retículo Endoplasmático/metabolismo , Humanos , Camundongos , Camundongos Knockout , Modelos Biológicos , Fosforilação , Traumatismo por Reperfusão/metabolismo , Fatores de Transcrição/metabolismo , eIF-2 Quinase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...