Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 211
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Neurochem ; 155(2): 191-206, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32157699

RESUMO

Mitochondrial dysfunction has a recognised role in the progression of Alzheimer's disease (AD) pathophysiology. Cerebral perfusion becomes increasingly inefficient throughout ageing, leading to unbalanced mitochondrial dynamics. This effect is exaggerated by amyloid ß (Aß) and phosphorylated tau, two hallmark proteins of AD pathology. A neuroprotective role for the adipose-derived hormone, leptin, has been demonstrated in neuronal cells. However, its effects with relation to mitochondrial function in AD remain largely unknown. To address this question, we have used both a glucose-serum-deprived (CGSD) model of ischaemic stroke in SH-SY5Y cells and a Aß1-42 -treatment model of AD in differentiated hippocampal cells. Using a combination of 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethylbenzimidazolylcarbocyanine iodide (JC-1) and MitoRed staining techniques, we show that leptin prevents depolarisation of the mitochondrial membrane and excessive mitochondrial fragmentation induced by both CGSD and Aß1-42 . Thereafter, we used ELISAs and a number of activity assays to reveal the biochemical underpinnings of these processes. Specifically, leptin was seen to inhibit up-regulation of the mitochondrial fission protein Fis1 and down-regulation of the mitochondrial fusion protein, Mfn2. Furthermore, leptin was seen to up-regulate the expression and activity of the antioxidant enzyme, monoamine oxidase B. Herein we provide the first demonstration that leptin is sufficient to protect against aberrant mitochondrial dynamics and resulting loss of function induced by both CGSD and Aß1-42 . We conclude that the established neuroprotective actions of leptin may be facilitated through regulation of mitochondrial dynamics.


Assuntos
Leptina/farmacologia , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Dinâmica Mitocondrial/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Peptídeos beta-Amiloides/farmacologia , Animais , Linhagem Celular , GTP Fosfo-Hidrolases/antagonistas & inibidores , GTP Fosfo-Hidrolases/biossíntese , Glucose/deficiência , Hipocampo/citologia , Hipocampo/patologia , Humanos , AVC Isquêmico/tratamento farmacológico , Camundongos , Mitocôndrias/efeitos dos fármacos , Membranas Mitocondriais/efeitos dos fármacos , Proteínas Mitocondriais/antagonistas & inibidores , Proteínas Mitocondriais/biossíntese , Monoaminoxidase/metabolismo , Fragmentos de Peptídeos/farmacologia , Espécies Reativas de Oxigênio/metabolismo
2.
Nucleic Acids Res ; 46(16): 8435-8453, 2018 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-30085276

RESUMO

Mammalian mitochondrial ribosomes (mitoribosomes) synthesize 13 proteins, essential components of the oxidative phosphorylation system. They are linked to mitochondrial disorders, often involving cardiomyopathy. Mitoribosome biogenesis is assisted by multiple cofactors whose specific functions remain largely uncharacterized. Here, we examined the role of human MTG1, a conserved ribosome assembly guanosine triphosphatase. MTG1-silencing in human cardiomyocytes and developing zebrafish revealed early cardiovascular lesions. A combination of gene-editing and biochemical approaches using HEK293T cells demonstrated that MTG1 binds to the large subunit (mtLSU) 16S ribosomal RNA to facilitate incorporation of late-assembly proteins. Furthermore, MTG1 interacts with mtLSU uL19 protein and mtSSU mS27, a putative guanosine triphosphate-exchange factor (GEF), to enable MTG1 release and the formation of the mB6 intersubunit bridge. In this way, MTG1 establishes a quality control checkpoint in mitoribosome assembly. In conclusion, MTG1 controls mitochondrial translation by coupling mtLSU assembly with intersubunit bridge formation using the intrinsic GEF activity acquired by the mtSSU through mS27, a unique occurrence in translational systems.


Assuntos
GTP Fosfo-Hidrolases/genética , Metaloproteínas/genética , Mitocôndrias/genética , Ribossomos Mitocondriais/metabolismo , Proteínas Nucleares/genética , Proteínas de Ligação a RNA/genética , Proteínas Ribossômicas/genética , Ribossomos/genética , Animais , Cardiomiopatias/genética , Cardiomiopatias/patologia , GTP Fosfo-Hidrolases/biossíntese , Inativação Gênica , Células HEK293 , Humanos , Mitocôndrias/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fosforilação Oxidativa , RNA Ribossômico 16S/genética , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento
3.
Hum Mol Genet ; 26(6): 1056-1069, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28087733

RESUMO

Alterations in mitochondrial activity and morphology have been demonstrated in human cells and tissues from individuals with Down syndrome (DS), as well as in DS mouse models. An impaired activity of the transcriptional coactivator PGC-1α/PPARGC1A due to the overexpression of chromosome 21 genes, such as NRIP1/RIP140, has emerged as an underlying cause of mitochondrial dysfunction in DS. We tested the hypothesis that the activation of the PGC-1α pathway might indeed reverse this mitochondrial dysfunction. To this end, we investigated the effects of metformin, a PGC-1α-activating drug, on mitochondrial morphology and function in DS foetal fibroblasts. Metformin induced both the expression of PGC-1α and an augmentation of its activity, as demonstrated by the increased expression of target genes, strongly promoting mitochondrial biogenesis. Furthermore, metformin enhanced oxygen consumption, ATP production, and overall mitochondrial activity. Most interestingly, this treatment reversed the fragmentation of mitochondria observed in DS and induced the formation of a mitochondrial network with a branched and elongated tubular morphology. Concomitantly, cristae remodelling occurred and the alterations observed by electron microscopy were significantly reduced. We finally demonstrated that the expression of genes of the fission/fusion machinery, namely OPA1 and MFN2, was reduced in trisomic cells and increased by metformin treatment. These results indicate that metformin promotes the formation of a mitochondrial network and corrects the mitochondrial dysfunction in DS cells. We speculate that alterations in the mitochondrial dynamics can be relevant in the pathogenesis of DS and that metformin can efficiently counteract these alterations, thus exerting protective effects against DS-associated pathologies.


Assuntos
Síndrome de Down/genética , Metformina/administração & dosagem , Mitocôndrias/efeitos dos fármacos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Trifosfato de Adenosina/biossíntese , Animais , Modelos Animais de Doenças , Síndrome de Down/tratamento farmacológico , Síndrome de Down/patologia , Fibroblastos/metabolismo , GTP Fosfo-Hidrolases/biossíntese , Humanos , Camundongos , Mitocôndrias/metabolismo , Dinâmica Mitocondrial/efeitos dos fármacos , Consumo de Oxigênio/efeitos dos fármacos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/biossíntese
4.
Circ Res ; 121(4): 392-410, 2017 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-28637784

RESUMO

RATIONALE: Mitochondria are important cellular organelles and play essential roles in maintaining cell structure and function. Emerging evidence indicates that in addition to having proinflammatory and proapoptotic effects, TNFα (tumor necrosis factor α) can, under certain circumstances, promote improvements in mitochondrial integrity and function, phenomena that can be ascribed to the existence of TNFR2 (TNFα receptor 2). OBJECTIVE: The present study aimed to investigate whether and how TNFR2 activation mediates the effects of TNFα on mitochondria. METHODS AND RESULTS: Freshly isolated neonatal mouse cardiac myocytes treated with shRNA targeting TNFR1 were used to study the effects of TNFR2 activation on mitochondrial function. Neonatal mouse cardiac myocytes exhibited increases in mitochondrial fusion, a change that was associated with increases in mitochondrial membrane potential, intracellular ATP levels, and oxygen consumption capacity. Importantly, TNFR2 activation-induced increases in OPA1 (optic atrophy 1) protein expression were responsible for the above enhancements, and these changes could be attenuated using siRNA targeting OPA1. Moreover, both Stat3 and RelA bound to the promoter region of OPA1 and their interactions synergistically upregulated OPA1 expression at the transcriptional level. Stat3 acetylation at lysine 370 or lysine 383 played a key role in the ability of Stat3 to form a supercomplex with RelA. Meanwhile, p300 modulated Stat3 acetylation in HEK293T (human embryonic kidney 293T) cells, and p300-mediated Stat3/RelA interactions played an indispensable role in OPA1 upregulation. Finally, TNFR2 activation exerted beneficial effects on OPA1 expression in an in vivo transverse aortic constriction model, whereby TNFR1-knockout mice exhibited better outcomes than in mice with both TNFR1 and TNFR2 knocked out. CONCLUSIONS: TNFR2 activation protects cardiac myocytes against stress by upregulating OPA1 expression. This process was facilitated by p300-mediated Stat3 acetylation and Stat3/RelA interactions, leading to improvements in mitochondrial morphology and function.


Assuntos
GTP Fosfo-Hidrolases/biossíntese , Dinâmica Mitocondrial/fisiologia , NF-kappa B/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Animais Recém-Nascidos , Doenças da Aorta/genética , Doenças da Aorta/metabolismo , Células Cultivadas , GTP Fosfo-Hidrolases/química , GTP Fosfo-Hidrolases/genética , Expressão Gênica , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/metabolismo , NF-kappa B/química , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Receptores Tipo II do Fator de Necrose Tumoral/química , Fator de Transcrição STAT3/química
5.
Circ Res ; 121(3): 220-233, 2017 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-28607103

RESUMO

RATIONALE: Mitochondrial changes occur during cell differentiation and cardiovascular disease. DRP1 (dynamin-related protein 1) is a key regulator of mitochondrial fission. We hypothesized that DRP1 plays a role in cardiovascular calcification, a process involving cell differentiation and a major clinical problem with high unmet needs. OBJECTIVE: To examine the effects of osteogenic promoting conditions on DRP1 and whether DRP1 inhibition alters the development of cardiovascular calcification. METHODS AND RESULTS: DRP1 was enriched in calcified regions of human carotid arteries, examined by immunohistochemistry. Osteogenic differentiation of primary human vascular smooth muscle cells increased DRP1 expression. DRP1 inhibition in human smooth muscle cells undergoing osteogenic differentiation attenuated matrix mineralization, cytoskeletal rearrangement, mitochondrial dysfunction, and reduced type 1 collagen secretion and alkaline phosphatase activity. DRP1 protein was observed in calcified human aortic valves, and DRP1 RNA interference reduced primary human valve interstitial cell calcification. Mice heterozygous for Drp1 deletion did not exhibit altered vascular pathology in a proprotein convertase subtilisin/kexin type 9 gain-of-function atherosclerosis model. However, when mineralization was induced via oxidative stress, DRP1 inhibition attenuated mouse and human smooth muscle cell calcification. Femur bone density was unchanged in mice heterozygous for Drp1 deletion, and DRP1 inhibition attenuated oxidative stress-mediated dysfunction in human bone osteoblasts. CONCLUSIONS: We demonstrate a new function of DRP1 in regulating collagen secretion and cardiovascular calcification, a novel area of exploration for the potential development of new therapies to modify cellular fibrocalcific response in cardiovascular diseases. Our data also support a role of mitochondrial dynamics in regulating oxidative stress-mediated arterial calcium accrual and bone loss.


Assuntos
GTP Fosfo-Hidrolases/antagonistas & inibidores , GTP Fosfo-Hidrolases/biossíntese , Proteínas Associadas aos Microtúbulos/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/biossíntese , Proteínas Mitocondriais/antagonistas & inibidores , Proteínas Mitocondriais/biossíntese , Miócitos de Músculo Liso/metabolismo , Estresse Oxidativo/fisiologia , Calcificação Vascular/metabolismo , Calcificação Vascular/prevenção & controle , Animais , Doenças das Artérias Carótidas/metabolismo , Doenças das Artérias Carótidas/patologia , Doenças das Artérias Carótidas/prevenção & controle , Células Cultivadas , Colágeno/metabolismo , Dinaminas , Humanos , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/patologia , Estresse Oxidativo/efeitos dos fármacos , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Calcificação Vascular/patologia
6.
Hum Mol Genet ; 25(14): 2972-2984, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27206984

RESUMO

Mutations in PARK2, encoding the E3 ubiquitin protein ligase Parkin, are a common cause of autosomal recessive Parkinson's disease (PD). Loss of PARK2 function compromises mitochondrial quality by affecting mitochondrial biogenesis, bioenergetics, dynamics, transport and turnover. We investigated the impact of PARK2 dysfunction on the endoplasmic reticulum (ER)-mitochondria interface, which mediates calcium (Ca2+) exchange between the two compartments and is essential for Parkin-dependent mitophagy. Confocal and electron microscopy analyses showed the ER and mitochondria to be in closer proximity in primary fibroblasts from PARK2 knockout (KO) mice and PD patients with PARK2 mutations than in controls. Ca2+ flux to the cytosol was also modified, due to enhanced ER-to-mitochondria Ca2+ transfers, a change that was also observed in neurons derived from induced pluripotent stem cells of a patient with PARK2 mutations. Subcellular fractionation showed the abundance of the Parkin substrate mitofusin 2 (Mfn2), which is known to modulate the ER-mitochondria interface, to be specifically higher in the mitochondrion-associated ER membrane compartment in PARK2 KO tissue. Mfn2 downregulation or the exogenous expression of normal Parkin restored cytosolic Ca2+ transients in fibroblasts from patients with PARK2 mutations. In contrast, a catalytically inactive PD-related Parkin variant had no effect. Overall, our data suggest that Parkin is directly involved in regulating ER-mitochondria contacts and provide new insight into the role of the loss of Parkin function in PD development.


Assuntos
Retículo Endoplasmático/metabolismo , GTP Fosfo-Hidrolases/genética , Mitocôndrias/metabolismo , Doença de Parkinson/genética , Ubiquitina-Proteína Ligases/genética , Animais , Sinalização do Cálcio/genética , Citosol/metabolismo , Retículo Endoplasmático/patologia , Fibroblastos , GTP Fosfo-Hidrolases/biossíntese , Regulação da Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Camundongos Knockout , Mitocôndrias/patologia , Mitofagia/genética , Mutação , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia
7.
Hum Mol Genet ; 25(19): 4266-4281, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27506976

RESUMO

Charcot-Marie-Tooth 2A (CMT2A) is an inherited peripheral neuropathy caused by mutations in MFN2, which encodes a mitochondrial membrane protein involved in mitochondrial network homeostasis. Because MFN2 is expressed ubiquitously, the reason for selective motor neuron (MN) involvement in CMT2A is unclear. To address this question, we generated MNs from induced pluripotent stem cells (iPSCs) obtained from the patients with CMT2A as an in vitro disease model. CMT2A iPSC-derived MNs (CMT2A-MNs) exhibited a global reduction in mitochondrial content and altered mitochondrial positioning without significant differences in survival and axon elongation. RNA sequencing profiles and protein studies of key components of the apoptotic executioner program (i.e. p53, BAX, caspase 8, cleaved caspase 3, and the anti-apoptotic marker Bcl2) demonstrated that CMT2A-MNs are more resistant to apoptosis than wild-type MNs. Exploring the balance between mitochondrial biogenesis and the regulation of autophagy-lysosome transcription, we observed an increased autophagic flux in CMT2A-MNs that was associated with increased expression of PINK1, PARK2, BNIP3, and a splice variant of BECN1 that was recently demonstrated to be a trigger for mitochondrial autophagic removal. Taken together, these data suggest that the striking reduction in mitochondria in MNs expressing mutant MFN2 is not the result of impaired biogenesis, but more likely the consequence of enhanced mitophagy. Thus, these pathways represent possible novel molecular therapeutic targets for the development of an effective cure for this disease.


Assuntos
Apoptose/genética , Doença de Charcot-Marie-Tooth/genética , GTP Fosfo-Hidrolases/genética , Proteínas Mitocondriais/genética , Neurônios Motores/metabolismo , Autofagia/genética , Proteína Beclina-1/genética , Doença de Charcot-Marie-Tooth/metabolismo , Doença de Charcot-Marie-Tooth/patologia , GTP Fosfo-Hidrolases/biossíntese , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Potencial da Membrana Mitocondrial/genética , Proteínas de Membrana/genética , Proteínas Mitocondriais/biossíntese , Neurônios Motores/patologia , Proteínas Quinases/genética , Proteínas Proto-Oncogênicas/genética , Ubiquitina-Proteína Ligases/genética
8.
PLoS Pathog ; 11(2): e1004629, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25658451

RESUMO

The ubiquitin proteasome system in plants plays important roles in plant-microbe interactions and in immune responses to pathogens. We previously demonstrated that the rice U-box E3 ligase SPL11 and its Arabidopsis ortholog PUB13 negatively regulate programmed cell death (PCD) and defense response. However, the components involved in the SPL11/PUB13-mediated PCD and immune signaling pathway remain unknown. In this study, we report that SPL11-interacting Protein 6 (SPIN6) is a Rho GTPase-activating protein (RhoGAP) that interacts with SPL11 in vitro and in vivo. SPL11 ubiquitinates SPIN6 in vitro and degrades SPIN6 in vivo via the 26S proteasome-dependent pathway. Both RNAi silencing in transgenic rice and knockout of Spin6 in a T-DNA insertion mutant lead to PCD and increased resistance to the rice blast pathogen Magnaporthe oryzae and the bacterial blight pathogen Xanthomonas oryzae pv. oryzae. The levels of reactive oxygen species and defense-related gene expression are significantly elevated in both the Spin6 RNAi and mutant plants. Strikingly, SPIN6 interacts with the small GTPase OsRac1, catalyze the GTP-bound OsRac1 into the GDP-bound state in vitro and has GAP activity towards OsRac1 in rice cells. Together, our results demonstrate that the RhoGAP SPIN6 acts as a linkage between a U-box E3 ligase-mediated ubiquitination pathway and a small GTPase-associated defensome system for plant immunity.


Assuntos
Morte Celular/imunologia , Proteínas Ativadoras de GTPase/metabolismo , Regulação da Expressão Gênica de Plantas/imunologia , Oryza/imunologia , Doenças das Plantas/imunologia , Imunidade Vegetal/imunologia , Apoptose/imunologia , GTP Fosfo-Hidrolases/biossíntese , GTP Fosfo-Hidrolases/imunologia , Imunidade Inata/imunologia , Imunoprecipitação , Proteínas de Plantas , Plantas Geneticamente Modificadas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Técnicas do Sistema de Duplo-Híbrido , Ubiquitina-Proteína Ligases/biossíntese , Ubiquitina-Proteína Ligases/imunologia , Ubiquitinação
9.
Stem Cells ; 34(3): 743-55, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26638184

RESUMO

Mouse skin mesenchymal stem cells (msMSCs) are dermis CD105(+) CD90(+) CD73(+) CD29(+) CD34(-) mesodermal precursors which, after in vitro induction, undergo chondro, adipo, and osteogenesis. Extensive metabolic reconfiguration has been found to occur during differentiation, and the bioenergetic status of a cell is known to be dependent on the quality and abundance of the mitochondrial population, which may be regulated by fusion and fission. However, little is known regarding the impact of mitochondrial dynamics on the differentiation process. We addressed this knowledge gap by isolating MSCs from Swiss female mice, inducing these cells to differentiate into osteo, chondro, and adipocytes and measuring changes in mass, morphology, dynamics, and bioenergetics. Mitochondrial biogenesis was increased in adipogenesis, as evaluated through confocal microscopy, citrate synthase activity, and mtDNA content. The early steps of adipo and osteogenesis involved mitochondrial elongation, as well as increased expression of mitochondrial fusion proteins Mfn1 and 2. Chondrogenesis involved a fragmented mitochondrial phenotype, increased expression of fission proteins Drp1, Fis1, and 2, and enhanced mitophagy. These events were accompanied by profound bioenergetic alterations during the commitment period. Moreover, knockdown of Mfn2 in adipo and osteogenesis and the overexpression of a dominant negative form of Drp1 during chondrogenesis resulted in a loss of differentiation ability. Overall, we find that mitochondrial morphology and its regulating processes of fission/fusion are modulated early on during commitment, leading to alterations in the bioenergetic profile that are important for differentiation. We thus propose a central role for mitochondrial dynamics in the maintenance/commitment of mesenchymal stem cells.


Assuntos
Diferenciação Celular/genética , Dinaminas/biossíntese , GTP Fosfo-Hidrolases/biossíntese , Células-Tronco Mesenquimais , Mitocôndrias/metabolismo , Adipogenia/genética , Animais , Condrogênese/genética , DNA Mitocondrial/genética , Dinaminas/genética , Feminino , GTP Fosfo-Hidrolases/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Camundongos , Mitocôndrias/genética , Dinâmica Mitocondrial/genética , Osteogênese/genética , Pele/citologia , Pele/metabolismo
10.
Biochim Biophys Acta ; 1849(12): 1423-31, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26518267

RESUMO

Excessive mitochondrial fission is associated with the pathogenesis of neurodegenerative diseases. Dynamin-related protein 1 (Drp1) possesses specific fission activity in the mitochondria and peroxisomes. Various post-translational modifications of Drp1 are known to modulate complex mitochondrial dynamics. However, the post-transcriptional regulation of Drp1 remains poorly understood. Here, we show that the heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1) regulates Drp1 expression at the post-transcriptional level. hnRNP A1 directly interacts with Drp1 mRNA at its 3'UTR region, and enhances translation potential without affecting mRNA stability. Down-regulation of hnRNP A1 induces mitochondrial elongation by reducing Drp1 expression. Moreover, depletion of hnRNP A1 suppresses 3-NP-mediated mitochondrial fission and dysfunction. In contrast, over-expression of hnRNP A1 promotes mitochondrial fragmentation by increasing Drp1 expression. Additionally, hnRNP A1 significantly exacerbates 3-NP-induced mitochondrial dysfunction and cell death in neuroblastoma cells. Interestingly, treatment with 3-NP induces subcellular translocation of hnRNP A1 from the nucleus to the cytoplasm, which accelerates the increase in Drp1 expression in hnRNP A1 over-expressing cells. Collectively, our findings suggest that hnRNP A1 controls mitochondrial dynamics by post-transcriptional regulation of Drp1.


Assuntos
GTP Fosfo-Hidrolases/biossíntese , Regulação Neoplásica da Expressão Gênica , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/fisiologia , Proteínas Associadas aos Microtúbulos/biossíntese , Dinâmica Mitocondrial/genética , Proteínas Mitocondriais/biossíntese , Proteínas de Neoplasias/fisiologia , Neuroblastoma/genética , Processamento Pós-Transcricional do RNA , Regiões 3' não Traduzidas/genética , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Dinaminas , GTP Fosfo-Hidrolases/genética , Ribonucleoproteína Nuclear Heterogênea A1 , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/genética , Humanos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Mitocondriais/genética , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Neuroblastoma/patologia , Ligação Proteica , Transporte Proteico , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Neoplásico/biossíntese , RNA Neoplásico/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA