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








Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 13(1): 4836, 2022 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-35977929

RESUMO

The mechanistic target of rapamycin (mTOR) signals through the mTOR complex 1 (mTORC1) and the mTOR complex 2 to maintain cellular and organismal homeostasis. Failure to finely tune mTOR activity results in metabolic dysregulation and disease. While there is substantial understanding of the molecular events leading mTORC1 activation at the lysosome, remarkably little is known about what terminates mTORC1 signaling. Here, we show that the AAA + ATPase Thorase directly binds mTOR, thereby orchestrating the disassembly and inactivation of mTORC1. Thorase disrupts the association of mTOR to Raptor at the mitochondria-lysosome interface and this action is sensitive to amino acids. Lack of Thorase causes accumulation of mTOR-Raptor complexes and altered mTORC1 disassembly/re-assembly dynamics upon changes in amino acid availability. The resulting excessive mTORC1 can be counteracted with rapamycin in vitro and in vivo. Collectively, we reveal Thorase as a key component of the mTOR pathway that disassembles and thus inhibits mTORC1.


Assuntos
Aminoácidos , Serina-Treonina Quinases TOR , ATPases Associadas a Diversas Atividades Celulares/metabolismo , Aminoácidos/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Fosforilação , Proteína Regulatória Associada a mTOR/metabolismo , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/metabolismo
2.
Cell Rep ; 33(5): 108329, 2020 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-33147468

RESUMO

The regulation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) trafficking affects multiple brain functions, such as learning and memory. We have previously shown that Thorase plays an important role in the internalization of AMPARs from the synaptic membrane. Here, we show that N-methyl-d-aspartate receptor (NMDAR) activation leads to increased S-nitrosylation of Thorase and N-ethylmaleimide-sensitive factor (NSF). S-nitrosylation of Thorase stabilizes Thorase-AMPAR complexes and enhances the internalization of AMPAR and interaction with protein-interacting C kinase 1 (PICK1). S-nitrosylated NSF is dependent on the S-nitrosylation of Thorase via trans-nitrosylation, which modulates the surface insertion of AMPARs. In the presence of the S-nitrosylation-deficient C137L Thorase mutant, AMPAR trafficking, long-term potentiation, and long-term depression are impaired. Overall, our data suggest that both S-nitrosylation and interactions of Thorase and NSF/PICK1 are required to modulate AMPAR-mediated synaptic plasticity. This study provides critical information that elucidates the mechanism underlying Thorase and NSF-mediated trafficking of AMPAR complexes.


Assuntos
ATPases Associadas a Diversas Atividades Celulares/metabolismo , Membrana Celular/metabolismo , Proteínas Sensíveis a N-Etilmaleimida/metabolismo , Receptores de AMPA/metabolismo , Adenosina Trifosfatases/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Ciclo Celular/metabolismo , Cisteína/metabolismo , Endocitose/efeitos dos fármacos , Glutationa/metabolismo , Células HEK293 , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout , N-Metilaspartato/farmacologia , Plasticidade Neuronal , Óxido Nítrico/metabolismo , Nitrosação , Ligação Proteica , Multimerização Proteica , Transporte Proteico , S-Nitrosoglutationa/metabolismo
3.
Cell Stem Cell ; 27(4): 633-645.e7, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32846140

RESUMO

The G2019S mutation in leucine-rich repeat kinase 2 (LRRK2) is a common cause of familial Parkinson's disease (PD). This mutation results in dopaminergic neurodegeneration via dysregulated protein translation, although how alterations in protein synthesis contribute to neurodegeneration in human neurons is not known. Here we define the translational landscape in LRRK2-mutant dopaminergic neurons derived from human induced pluripotent stem cells (hiPSCs) via ribosome profiling. We found that mRNAs that have complex secondary structure in the 5' untranslated region (UTR) are translated more efficiently in G2019S LRRK2 neurons. This leads to the enhanced translation of multiple genes involved in Ca2+ regulation and to increased Ca2+ influx and elevated intracellular Ca2+ levels, a major contributor to PD pathogenesis. This study reveals a link between dysregulated translation control and Ca2+ homeostasis in G2019S LRRK2 human dopamine neurons, which potentially contributes to the progressive and selective dopaminergic neurotoxicity in PD.


Assuntos
Células-Tronco Pluripotentes Induzidas , Doença de Parkinson , Cálcio , Neurônios Dopaminérgicos/metabolismo , Homeostase , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Mutação/genética , Doença de Parkinson/genética , Biossíntese de Proteínas
4.
J Neurosci ; 40(32): 6234-6249, 2020 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-32605938

RESUMO

The G2019S mutation in leucine-rich repeat kinase 2 (LRRK2) is a common cause of Parkinson's disease (PD) and results in age-related dopamine neuron loss and locomotor dysfunction in Drosophila melanogaster through an aberrant increase in bulk neuronal protein synthesis. Under nonpathologic conditions, protein synthesis is tightly controlled by metabolic regulation. Whether nutritional and metabolic influences on protein synthesis can modulate the pathogenic effect of LRRK2 on protein synthesis and thereby impact neuronal loss is a key unresolved question. Here, we show that LRRK2 G2019S-induced neurodegeneration is critically dependent on dietary amino acid content in Drosophila studies with both sexes. Low dietary amino acid concentration prevents aberrant protein synthesis and blocks LRRK2 G2019S-mediated neurodegeneration in Drosophila and rat primary neurons. Unexpectedly, a moderately high-amino acid diet also blocks dopamine neuron loss and motor deficits in Drosophila through a separate mechanism involving stress-responsive activation of 5'-AMP-activated protein kinase (AMPK) and neuroprotective induction of autophagy, implicating the importance of protein homeostasis to neuronal viability. At the highest amino acid diet of the range tested, PD-related neurodegeneration occurs in an age-related manner, but is also observed in control strains, suggesting that it is independent of mutant LRRK2 expression. We propose that dietary influences on protein synthesis and autophagy are critical determinants of LRRK2 neurodegeneration, opening up possibilities for future therapeutic intervention.SIGNIFICANCE STATEMENT Parkinson's disease (PD) prevalence is projected to rise as populations continue to age, yet there are no current therapeutic approaches that delay or stop disease progression. A broad role for leucine-rich repeat kinase 2 (LRRK2) mutations in familial and idiopathic PD has emerged. Here, we show that dietary amino acids are important determinants of neurodegeneration in a Drosophila model of LRRK2 PD. Restricting all amino acids effectively suppresses dopaminergic neuron loss and locomotor deficits and is associated with reduced protein synthesis, while moderately high amino acids similarly attenuate these PD-related phenotypes through a stress-responsive induction of 5'-AMP-activated protein kinase and autophagy. These studies suggest that diet plays an important role in the development of PD-related phenotypes linked to LRRK2.


Assuntos
Aminoácidos/farmacologia , Autofagia , Neurônios Dopaminérgicos/metabolismo , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Doença de Parkinson/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Aminoácidos/administração & dosagem , Aminoácidos/metabolismo , Animais , Células Cultivadas , Dietoterapia/métodos , Neurônios Dopaminérgicos/efeitos dos fármacos , Drosophila , Feminino , Masculino , Doença de Parkinson/dietoterapia , Doença de Parkinson/genética , Proteínas Quinases/metabolismo , Ratos , Ratos Sprague-Dawley
6.
Brain ; 141(3): 651-661, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29390050

RESUMO

Members of the AAA+ superfamily of ATPases are involved in the unfolding of proteins and disassembly of protein complexes and aggregates. ATAD1 encoding the ATPase family, AAA+ domain containing 1-protein Thorase plays an important role in the function and integrity of mitochondria and peroxisomes. Postsynaptically, Thorase controls the internalization of excitatory, glutamatergic AMPA receptors by disassembling complexes between the AMPA receptor-binding protein, GRIP1, and the AMPA receptor subunit GluA2. Using whole-exome sequencing, we identified a homozygous frameshift mutation in the last exon of ATAD1 [c.1070_1071delAT; p.(His357Argfs*15)] in three siblings who presented with a severe, lethal encephalopathy associated with stiffness and arthrogryposis. Biochemical and cellular analyses show that the C-terminal end of Thorase mutant gained a novel function that strongly impacts its oligomeric state, reduces stability or expression of a set of Golgi, peroxisomal and mitochondrial proteins and affects disassembly of GluA2 and Thorase oligomer complexes. Atad1-/- neurons expressing Thorase mutantHis357Argfs*15 display reduced amount of GluA2 at the cell surface suggesting that the Thorase mutant may inhibit the recycling back and/or reinsertion of AMPA receptors to the plasma membrane. Taken together, our molecular and functional analyses identify an activating ATAD1 mutation as a new cause of severe encephalopathy and congenital stiffness.


Assuntos
ATPases Associadas a Diversas Atividades Celulares/genética , Encefalopatias/genética , Regulação da Expressão Gênica/genética , Mutação/genética , Neurônios/patologia , Receptores de AMPA/metabolismo , Adenosina Trifosfatases/metabolismo , Encefalopatias/diagnóstico por imagem , Encefalopatias/patologia , Proteínas de Transporte/metabolismo , Análise Mutacional de DNA , Saúde da Família , Feminino , Homozigoto , Humanos , Lactente , Imageamento por Ressonância Magnética , Masculino , Mitocôndrias/genética , Mitocôndrias/patologia , Modelos Moleculares , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Neurônios/ultraestrutura , Consumo de Oxigênio/genética , Transporte Proteico/genética , RNA Mensageiro/metabolismo
7.
Mol Ther ; 23(3): 570-7, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25418680

RESUMO

Efficient and precise genome editing is crucial for realizing the full research and therapeutic potential of human induced pluripotent stem cells (iPSCs). Engineered nucleases including CRISPR/Cas9 and transcription activator like effector nucleases (TALENs) provide powerful tools for enhancing gene-targeting efficiency. In this study, we investigated the relative efficiencies of CRISPR/Cas9 and TALENs in human iPSC lines for inducing both homologous donor-based precise genome editing and nonhomologous end joining (NHEJ)-mediated gene disruption. Significantly higher frequencies of NHEJ-mediated insertions/deletions were detected at several endogenous loci using CRISPR/Cas9 than using TALENs, especially at nonexpressed targets in iPSCs. In contrast, comparable efficiencies of inducing homologous donor-based genome editing were observed at disease-associated loci in iPSCs. In addition, we investigated the specificity of guide RNAs used in the CRISPR/Cas9 system in targeting disease-associated point mutations in patient-specific iPSCs. Using myeloproliferative neoplasm patient-derived iPSCs that carry an acquired JAK2-V617F point mutation and α1-antitrypsin (AAT) deficiency patient-derived iPSCs that carry an inherited Z-AAT point mutation, we demonstrate that Cas9 can specifically target either the mutant or the wild-type allele with little disruption at the other allele differing by a single nucleotide. Overall, our results demonstrate the advantages of the CRISPR/Cas9 system in allele-specific genome targeting and in NHEJ-mediated gene disruption.


Assuntos
Alelos , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Genoma Humano , Células-Tronco Pluripotentes Induzidas/metabolismo , Locos de Características Quantitativas , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sequência de Bases , Proteína 9 Associada à CRISPR , Linhagem Celular , Reparo do DNA por Junção de Extremidades , Endonucleases/genética , Endonucleases/metabolismo , Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Dados de Sequência Molecular , Mutação , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/metabolismo , Transtornos Mieloproliferativos/patologia , RNA Guia de Cinetoplastídeos/genética , RNA Guia de Cinetoplastídeos/metabolismo , Reparo de DNA por Recombinação , alfa 1-Antitripsina/genética , alfa 1-Antitripsina/metabolismo
8.
Fly (Austin) ; 8(3): 165-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25483009

RESUMO

LRRK2 mutations are a frequent cause of familial Parkinson disease (PD) and are also found in a number of sporadic PD cases. PD-linked G2019S and I2020T mutations in the kinase domain of LRRK2 result in elevated kinase activity, which is required for the toxicity of these pathogenic variants in cell and animal models of PD. We recently reported that LRRK2 interacts with and phosphorylates a number of mammalian ribosomal proteins, several of which exhibit increased phosphorylation via both G2019S and I2020T LRRK2. Blocking the phosphorylation of ribosomal protein s15 through expression of phospho-deficient T136A s15 prevents age-associated locomotor deficits and dopamine neuron loss caused by G2019S LRRK2 expression in Drosophila indicating that s15 is a pathogenic LRRK2 substrate. We previously described that G2019S LRRK2 causes an induction of bulk mRNA translation that is blocked by T136A s15 or the protein synthesis inhibitor anisomycin. Here, we report the protective effects of the eIF4E/eIF4G interaction inhibitor 4EGI-1, in preventing neurodegenerative phenotypes in G2019S LRRK2 flies, and discuss how our findings and those of other groups provide a framework to begin investigating the mechanistic impact of LRRK2 on translation.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Hidrazonas/uso terapêutico , Doença de Parkinson/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Ribossômicas/metabolismo , Tiazóis/uso terapêutico , Animais , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Hidrazonas/farmacologia , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Doença de Parkinson/genética , Doença de Parkinson/prevenção & controle , Fenótipo , Biossíntese de Proteínas/efeitos dos fármacos , Tiazóis/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA