Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 148(4): 716-26, 2012 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-22341444

RESUMO

Mitochondrial dysfunction causes poorly understood tissue-specific pathology stemming from primary defects in respiration, coupled with altered reactive oxygen species (ROS), metabolic signaling, and apoptosis. The A1555G mtDNA mutation that causes maternally inherited deafness disrupts mitochondrial ribosome function, in part, via increased methylation of the mitochondrial 12S rRNA by the methyltransferase mtTFB1. In patient-derived A1555G cells, we show that 12S rRNA hypermethylation causes ROS-dependent activation of AMP kinase and the proapoptotic nuclear transcription factor E2F1. This retrograde mitochondrial-stress relay is operative in vivo, as transgenic-mtTFB1 mice exhibit enhanced 12S rRNA methylation in multiple tissues, increased E2F1 and apoptosis in the stria vascularis and spiral ganglion neurons of the inner ear, and progressive E2F1-dependent hearing loss. This mouse mitochondrial disease model provides a robust platform for deciphering the complex tissue specificity of human mitochondrial-based disorders, as well as the precise pathogenic mechanism of maternally inherited deafness and its exacerbation by environmental factors.


Assuntos
Surdez/metabolismo , Modelos Animais de Doenças , Fator de Transcrição E2F1/metabolismo , Animais , DNA Mitocondrial/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Orelha Interna/patologia , Cistos Glanglionares/patologia , Humanos , Camundongos , Camundongos Transgênicos , Mitocôndrias/metabolismo , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Mutação , Neurônios/patologia , RNA Ribossômico/metabolismo , Espécies Reativas de Oxigênio , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
2.
J Interprof Care ; 35(4): 532-537, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32917104

RESUMO

Musculoskeletal pain is a prominent complaint in primary care resulting in increased referrals to physical therapy (PT); however, the referral system often results in delays and discontinuation of care. Several models have been developed to improve the referral process including integrating PT into primary care clinics. The Veterans Health Administration (VHA) Center of Excellence in Primary Care Education (CoEPCE), which educates post-graduate trainees in interprofessional teams, began (in 2015) embedding physical therapists into primary care clinics enabling patients to see a physical therapist during their primary care visit. To evaluate the efficacy of this model we tracked the numbers of PT referrals, the number of completed referrals, and the length of time between referral and completion. PT referral parameters from PT-integrated trainees in the CoEPCE were compared to two traditional primary care training clinics at the same VHA site (Firm A and Firm B). Results indicate that the CoEPCE placed and completed more PT referrals and did so with a shorter turnaround time than was seen in the other two clinics. Further analysis suggests that the decreased turnaround time can be attributed to the integration of PTs into the primary care clinic. The results support extending the use of interprofessional clinics that integrate PT into primary care settings.


Assuntos
Relações Interprofissionais , Atenção Primária à Saúde , Instituições de Assistência Ambulatorial , Humanos , Modalidades de Fisioterapia , Encaminhamento e Consulta
3.
Am J Pathol ; 185(12): 3132-40, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26552864

RESUMO

The A1555G mutation in the 12S rRNA gene of human mitochondrial DNA causes maternally inherited, nonsyndromic deafness, an extreme case of tissue-specific mitochondrial pathology. A transgenic mouse strain that robustly overexpresses the mitochondrial 12S ribosomal RNA methyltransferase TFB1M (Tg-mtTFB1 mice) exhibits progressive hearing loss that we proposed models aspects of A1555G-related pathology in humans. Although our previous studies of Tg-mtTFB1 mice implicated apoptosis in the spiral ganglion and stria vascularis because of mitochondrial reactive oxygen species-mediated activation of AMP kinase (AMPK) and the nuclear transcription factor E2F1, detailed auditory pathology was not delineated. Herein, we show that Tg-mtTFB1 mice have reduced endocochlear potential, indicative of significant stria vascularis dysfunction, but without obvious signs of strial atrophy. We also observed decreased auditory brainstem response peak 1 amplitude and prolonged wave I latency, consistent with apoptosis of spiral ganglion neurons. Although no major loss of hair cells was observed, there was a mild impairment of voltage-dependent electromotility of outer hair cells. On the basis of these results, we propose that these events conspire to produce the progressive hearing loss phenotype in Tg-mtTFB1 mice. Finally, genetically reducing AMPK α1 rescues hearing loss in Tg-mtTFB1 mice, confirming that aberrant up-regulation of AMPK signaling promotes the observed auditory pathology. The relevance of these findings to human A1555G patients and the potential therapeutic value of reducing AMPK activity are discussed.


Assuntos
Surdez/patologia , Doenças Mitocondriais/patologia , Proteínas Quinases Ativadas por AMP/deficiência , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/fisiologia , Animais , Apoptose/fisiologia , DNA Mitocondrial/genética , Surdez/genética , Surdez/fisiopatologia , Modelos Animais de Doenças , Potenciais Evocados Auditivos do Tronco Encefálico/fisiologia , Células Ciliadas Auditivas Internas/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Doenças Mitocondriais/genética , Doenças Mitocondriais/fisiopatologia , Mutação , Órgão Espiral/patologia , Tempo de Reação/fisiologia , Transdução de Sinais/fisiologia , Gânglio Espiral da Cóclea/patologia , Gânglio Espiral da Cóclea/fisiopatologia , Estria Vascular/patologia , Fatores de Transcrição/genética
4.
Hum Mol Genet ; 20(22): 4452-61, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21862453

RESUMO

Germline mutations in SDHD, a mitochondrial complex II (succinate dehydrogenase) subunit gene at chromosome band 11q23, cause highly penetrant paraganglioma (PGL) tumors when transmitted through fathers. In contrast, maternal transmission rarely, if ever, leads to tumor development. The mechanism underlying this unusual monogenic tumor predisposition pattern is poorly understood. Here, we describe identification of imprinted methylation within an alternative promoter for a large intergenic non-coding RNA located at a distant gene desert boundary flanking SDHD. Methylation at this site primarily occurs within two consecutive HpaII restriction enzyme sites in a tissue-specific manner, most commonly in the adrenal gland. Informative fetal tissues and PGL tumors demonstrate maternal allelic hypermethylation. While a strong binding site for the enhancer-blocking protein CTCF within the alternative promoter shows no evidence of methylation, hyper-methylated adrenal tissues show increased binding of the chromatin-looping factor cohesin relative to the hypo-methylated tissues. These results suggest that the differential allelic methylation we observe at this locus is associated with altered chromatin architectures. These results provide molecular evidence for imprinting at a boundary element flanking the SDHD locus and suggest that epigenetic suppression of the maternal allele is the underlying mechanism of the imprinted penetrance of SDHD mutations.


Assuntos
Impressão Genômica/genética , Elementos Isolantes/genética , Succinato Desidrogenase/genética , Imunoprecipitação da Cromatina , Biologia Computacional , Metilação de DNA/genética , Humanos , Técnicas In Vitro , Mutação , Polimorfismo Genético/genética
5.
Am J Pathol ; 178(6): 2740-51, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21641396

RESUMO

Immune system-related pathology is common in ataxia-telangiectasia (A-T) patients and mice that lack the protein kinase, A-T mutated (ATM). However, it has not been studied how ATM influences immune responses to a viral infection. Using the lymphocytic choriomeningitis virus (LCMV) infection model, we show that ATM(-/-) mice, despite having fewer naïve CD8⁺ T cells, effectively clear the virus. However, aberrant CD8⁺ T-cell responses are observed, including defective expansion and contraction, effector-to-memory differentiation, and a switch in viral-epitope immunodominance. T-cell receptor-activated, but not naïve, ATM(-/-) splenic CD8⁺ T cells have increased ribosomal protein S6 and Akt phosphorylation and do not proliferate well in response to IL-15, a cytokine important for memory T-cell development. Accordingly, pharmacological Akt or mammalian target of rapamycin complex 1 (mTORC1) inhibition during T-cell receptor activation alone rescues the IL-15 proliferation defect. Finally, rapamycin treatment during LCMV infection in vivo increases the number of memory T cells in ATM(-/-) mice. Altogether, these results show that CD8⁺T cells lacking ATM have hyperactive Akt and mTORC1 signaling in response to T-cell receptor activation, which results in aberrant cytokine responses and memory T-cell development. We speculate that similar signaling defects contribute to the immune system pathology of A-T, and that inhibition of Akt and/or mTORC1 may be of therapeutic value.


Assuntos
Ataxia Telangiectasia/virologia , Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular/imunologia , Memória Imunológica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Ataxia Telangiectasia/complicações , Ataxia Telangiectasia/imunologia , Proteínas Mutadas de Ataxia Telangiectasia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/enzimologia , Linfócitos T CD8-Positivos/patologia , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Ativação Enzimática/efeitos dos fármacos , Memória Imunológica/efeitos dos fármacos , Interleucina-15/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Coriomeningite Linfocítica/complicações , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/efeitos dos fármacos , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Complexos Multiproteicos , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Serina-Treonina Quinases TOR , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/metabolismo
6.
Hum Mol Genet ; 18(14): 2670-82, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19417006

RESUMO

Mitochondrial biogenesis is controlled by signaling networks that relay information to and from the organelles. However, key mitochondrial factors that mediate such pathways and how they contribute to human disease are not understood fully. Here we demonstrate that the rRNA methyltransferase-related human mitochondrial transcription factors B1 and B2 are key downstream effectors of mitochondrial biogenesis that perform unique, yet cooperative functions. The primary function of h-mtTFB2 is mtDNA transcription and maintenance, which is independent of its rRNA methyltransferase activity, while that of h-mtTFB1 is mitochondrial 12S rRNA methylation needed for normal mitochondrial translation, metabolism and cell growth. Over-expression of h-mtTFB1 causes 12S rRNA hypermethylation, aberrant mitochondrial biogenesis and increased sorbitol-induced cell death. These phenotypes are recapitulated in cells harboring the pathogenic A1555G mtDNA mutation, implicating a deleterious rRNA methylation-dependent retrograde signal in maternally inherited deafness pathology and shedding significant insight into how h-mtTFB1 acts as a nuclear modifier of this disease.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Surdez/enzimologia , Impressão Genômica , Metiltransferases/metabolismo , Mitocôndrias/enzimologia , Proteínas Mitocondriais/metabolismo , Fatores de Transcrição/metabolismo , tRNA Metiltransferases/metabolismo , Linhagem Celular , Proteínas de Ligação a DNA/genética , Surdez/genética , Surdez/metabolismo , Humanos , Metilação , Metiltransferases/genética , Mitocôndrias/genética , Mitocôndrias/metabolismo , Proteínas Mitocondriais/genética , RNA Ribossômico/genética , RNA Ribossômico/metabolismo , Fatores de Transcrição/genética , tRNA Metiltransferases/genética
7.
Cell Metab ; 28(5): 776-786.e5, 2018 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-30122556

RESUMO

Transient mitochondrial stress can promote beneficial physiological responses and longevity, termed "mitohormesis." To interrogate mitohormetic pathways in mammals, we generated mice in which mitochondrial superoxide dismutase 2 (SOD2) can be knocked down in an inducible and reversible manner (iSOD2-KD mice). Depleting SOD2 only during embryonic development did not cause post-natal lethality, allowing us to probe adaptive responses to mitochondrial oxidant stress in adult mice. Liver from adapted mice had increased mitochondrial biogenesis and antioxidant gene expression and fewer reactive oxygen species. Gene expression analysis implicated non-canonical activation of the Nrf2 antioxidant and PPARγ/PGC-1α mitochondrial signaling pathways in this response. Transient SOD2 knockdown in embryonic fibroblasts from iSOD2-KD mice also resulted in adaptive mitochondrial changes, enhanced antioxidant capacity, and resistance to a subsequent oxidant challenge. We propose that mitohormesis in response to mitochondrial oxidative stress in mice involves sustained activation of mitochondrial and antioxidant signaling pathways to establish a heightened basal antioxidant state.


Assuntos
Mitocôndrias/metabolismo , Estresse Oxidativo , Transdução de Sinais , Superóxido Dismutase/metabolismo , Animais , Antioxidantes/metabolismo , Feminino , Longevidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/genética
8.
J Biol Chem ; 283(16): 10632-41, 2008 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-18276591

RESUMO

The Kv2.1 gene encodes a highly conserved delayed rectifier potassium channel that is widely expressed in neurons of the central nervous system. In the bag cell neurons of Aplysia, Kv2.1 channels contribute to the repolarization of action potentials during a prolonged afterdischarge that triggers a series of reproductive behaviors. Partial inactivation of Aplysia Kv2.1 during repetitive firing produces frequency-dependent broadening of action potentials during the afterdischarge. We have now found that, as in mammalian neurons, Kv2.1 channels in bag cell neurons are localized to ring-like clusters in the plasma membrane of the soma and proximal dendrites. Either elevation of cyclic AMP levels or direct electrical stimulation of afterdischarge rapidly enhanced formation of these clusters on the somata of these neurons. In contrast, injection of a 13-amino acid peptide corresponding to a region in the C terminus that is required for clustering of Kv2.1 channels produced disassociation of the clusters, resulting in a more uniform distribution over the somata. Voltage clamp recordings demonstrated that peptide-induced dissociation of the Kv2.1 clusters is associated with an increase in the amplitude of delayed rectifier current and a shift of activation toward more negative potentials. In current clamp recording, injection of the unclustering peptide reduced the width of action potentials and reduced frequency-dependent broadening of action potentials. Our results suggest that rapid redistribution of Kv2.1 channels occurs during physiological changes in neuronal excitability.


Assuntos
Aplysia/metabolismo , Neurônios/metabolismo , Canais de Potássio Shab/metabolismo , Transmissão Sináptica , Potenciais de Ação , Animais , Células CHO , Membrana Celular/metabolismo , Células Cultivadas , Cricetinae , Cricetulus , Potenciais da Membrana , Modelos Biológicos , Técnicas de Patch-Clamp , Peptídeos/química , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA