Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
J Neurosci ; 39(4): 678-691, 2019 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-30504273

RESUMO

The actin cytoskeleton is crucial for neuronal migration in the mammalian developing cerebral cortex. The adaptor protein Drebrin-like (Dbnl) plays important roles in reorganization of the actin cytoskeleton, dendrite formation, and endocytosis by interacting with F-actin, cobl, and dynamin. Although Dbnl is known to be expressed in the brain, the functions of this molecule during brain development are largely unknown. In this study, to examine the roles of Dbnl in the developing cerebral cortex, we conducted experiments using mice of both sexes with knockdown of Dbnl, effected by in utero electroporation, in the migrating neurons of the embryonic cortex. Time-lapse imaging of the Dbnl-knockdown neurons revealed that the presence of Dbnl is a prerequisite for appropriate formation of processes in the multipolar neurons in the multipolar cell accumulation zone or the deep part of the subventricular zone, and for neuronal polarization and entry into the cortical plate. We found that Dbnl knockdown decreased the amount of N-cadherin protein expressed on the plasma membrane of the cortical neurons. The defect in neuronal migration caused by Dbnl knockdown was rescued by moderate overexpression of N-cadherin and αN-catenin or by transfection of the phospho-mimic form (Y337E, Y347E), but not the phospho-resistant form (Y337F, Y347F), of Dbnl. These results suggest that Dbnl controls neuronal migration, neuronal multipolar morphology, and cell polarity in the developing cerebral cortex via regulating N-cadherin expression.SIGNIFICANCE STATEMENT Disruption of neuronal migration can cause neuronal disorders, such as lissencephaly and subcortical band heterotopia. During cerebral cortical development, the actin cytoskeleton plays a key role in neuronal migration; however, the mechanisms of regulation of neuronal migration by the actin cytoskeleton still remain unclear. Herein, we report that the novel protein Dbnl, an actin-binding protein, controls multiple events during neuronal migration in the developing mouse cerebral cortex. We also showed that this regulation is mediated by phosphorylation of Dbnl at tyrosine residues 337 and 347 and αN-catenin/N-cadherin, suggesting that the Dbnl-αN-catenin/N-cadherin pathway is important for neuronal migration in the developing cortex.


Assuntos
Caderinas/biossíntese , Movimento Celular/fisiologia , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/fisiologia , Proteínas dos Microfilamentos/fisiologia , Neurônios/fisiologia , Domínios de Homologia de src/fisiologia , Animais , Caderinas/genética , Membrana Celular/metabolismo , Córtex Cerebral/embriologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Ventrículos Laterais/citologia , Ventrículos Laterais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos ICR , Proteínas dos Microfilamentos/biossíntese , Proteínas dos Microfilamentos/genética , Neurônios/ultraestrutura , Gravidez , Domínios de Homologia de src/genética
2.
Mol Psychiatry ; 23(10): 2090-2110, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30283027

RESUMO

Early-phase pathologies of Alzheimer's disease (AD) are attracting much attention after clinical trials of drugs designed to remove beta-amyloid (Aß) aggregates failed to recover memory and cognitive function in symptomatic AD patients. Here, we show that phosphorylation of serine/arginine repetitive matrix 2 (SRRM2) at Ser1068, which is observed in the brains of early phase AD mouse models and postmortem end-stage AD patients, prevents its nuclear translocation by inhibiting interaction with T-complex protein subunit α. SRRM2 deficiency in neurons destabilized polyglutamine binding protein 1 (PQBP1), a causative gene for intellectual disability (ID), greatly affecting the splicing patterns of synapse-related genes, as demonstrated in a newly generated PQBP1-conditional knockout model. PQBP1 and SRRM2 were downregulated in cortical neurons of human AD patients and mouse AD models, and the AAV-PQBP1 vector recovered RNA splicing, the synapse phenotype, and the cognitive decline in the two mouse models. Finally, the kinases responsible for the phosphorylation of SRRM2 at Ser1068 were identified as ERK1/2 (MAPK3/1). These results collectively reveal a new aspect of AD pathology in which a phosphorylation signal affecting RNA splicing and synapse integrity precedes the formation of extracellular Aß aggregates and may progress in parallel with tau phosphorylation.


Assuntos
Doença de Alzheimer/genética , Proteínas de Transporte/genética , Proteínas Nucleares/genética , Proteínas de Ligação a RNA/genética , Transporte Ativo do Núcleo Celular , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/metabolismo , Proteínas de Transporte/metabolismo , Cognição , Proteínas de Ligação a DNA , Modelos Animais de Doenças , Humanos , Células-Tronco Pluripotentes Induzidas , Deficiência Intelectual/genética , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Fosforilação , Cultura Primária de Células , Splicing de RNA , Proteínas de Ligação a RNA/metabolismo , Proteínas tau/metabolismo
3.
Hum Mol Genet ; 25(20): 4432-4447, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-28173122

RESUMO

DNA damage and repair is a critical domain of many neurodegenerative diseases. In this study, we focused on RpA1, a candidate key molecule in polyQ disease pathologies, and tested the therapeutic effect of adeno-associated virus (AAV) vector expressing RpA1 on mutant Ataxin-1 knock-in (Atxn1-KI) mice. We found significant effects on motor functions, normalized DNA damage markers (γH2AX and 53BP1), and improved Purkinje cell morphology; effects that lasted for 50 weeks following AAV-RpA1 infection. In addition, we confirmed that AAV-RpA1 indirectly recovered multiple cellular functions such as RNA splicing, transcription and cell cycle as well as abnormal morphology of dendrite and dendritic spine of Purkinje cells in Atxn1-KI mice. All these results suggested a possibility of gene therapy with RpA1 for SCA1.


Assuntos
Ataxina-1/genética , Reparo do DNA , Mutação , Proteína de Replicação A/metabolismo , Ataxias Espinocerebelares/metabolismo , Animais , Ciclo Celular , DNA/metabolismo , Dano ao DNA , Dependovirus , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Terapia Genética , Camundongos , Células de Purkinje/metabolismo , Células de Purkinje/patologia , Células de Purkinje/fisiologia , RNA/metabolismo , Splicing de RNA , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/patologia , Ataxias Espinocerebelares/fisiopatologia , Transcrição Gênica
4.
Hum Mol Genet ; 25(21): 4749-4770, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28171658

RESUMO

Neuronal cell death in neurodegenerative diseases is not fully understood. Here we report that mutant huntingtin (Htt), a causative gene product of Huntington's diseases (HD) selectively induces a new form of necrotic cell death, in which endoplasmic reticulum (ER) enlarges and cell body asymmetrically balloons and finally ruptures. Pharmacological and genetic analyses revealed that the necrotic cell death is distinct from the RIP1/3 pathway-dependent necroptosis, but mediated by a functional deficiency of TEAD/YAP-dependent transcription. In addition, we revealed that a cell cycle regulator, Plk1, switches the balance between TEAD/YAP-dependent necrosis and p73/YAP-dependent apoptosis by shifting the interaction partner of YAP from TEAD to p73 through YAP phosphorylation at Thr77. In vivo ER imaging with two-photon microscopy detects similar ER enlargement, and viral vector-mediated delivery of YAP as well as chemical inhibitors of the Hippo pathway such as S1P recover the ER instability and necrosis in HD model mice. Intriguingly S1P completely stops the decline of motor function of HD model mice even after the onset of symptom. Collectively, we suggest approaches targeting the signalling pathway of TEAD/YAP-transcription-dependent necrosis (TRIAD) could lead to a therapeutic development against HD.


Assuntos
Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Necrose/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proteínas de Ciclo Celular , Morte Celular , Proteínas de Ligação a DNA/genética , Retículo Endoplasmático/metabolismo , Humanos , Doença de Huntington/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Necrose/genética , Neurônios/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Fosforilação , Cultura Primária de Células , Ligação Proteica , Transdução de Sinais , Transativadores/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
5.
Hum Mol Genet ; 24(2): 540-58, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25231903

RESUMO

Using a high-end mass spectrometry, we screened phosphoproteins and phosphopeptides in four types of Alzheimer's disease (AD) mouse models and human AD postmortem brains. We identified commonly changed phosphoproteins in multiple models and also determined phosphoproteins related to initiation of amyloid beta (Aß) deposition in the mouse brain. After confirming these proteins were also changed in and human AD brains, we put the proteins on experimentally verified protein-protein interaction databases. Surprisingly, most of the core phosphoproteins were directly connected, and they formed a functional network linked to synaptic spine formation. The change of the core network started at a preclinical stage even before histological Aß deposition. Systems biology analyses suggested that phosphorylation of myristoylated alanine-rich C-kinase substrate (MARCKS) by overactivated kinases including protein kinases C and calmodulin-dependent kinases initiates synapse pathology. Two-photon microscopic observation revealed recovery of abnormal spine formation in the AD model mice by targeting a core protein MARCKS or by inhibiting candidate kinases, supporting our hypothesis formulated based on phosphoproteome analysis.


Assuntos
Doença de Alzheimer/metabolismo , Fosfoproteínas/metabolismo , Sinapses/metabolismo , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Substrato Quinase C Rico em Alanina Miristoilada , Fosfoproteínas/genética , Fosforilação , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Transdução de Sinais
6.
Hum Mol Genet ; 23(5): 1345-64, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24179173

RESUMO

DNA damage repair is implicated in neurodegenerative diseases; however, the relative contributions of various DNA repair systems to the pathology of these diseases have not been investigated systematically. In this study, we performed a systematic in vivo screen of all available Drosophila melanogaster homolog DNA repair genes, and we tested the effect of their overexpression on lifespan and developmental viability in Spinocerebellar Ataxia Type 1 (SCA1) Drosophila models expressing human mutant Ataxin-1 (Atxn1). We identified genes previously unknown to be involved in CAG-/polyQ-related pathogenesis that function in multiple DNA damage repair systems. Beyond the significance of each repair system, systems biology analyses unraveled the core networks connecting positive genes in the gene screen that could contribute to SCA1 pathology. In particular, RpA1, which had the largest effect on lifespan in the SCA1 fly model, was located at the hub position linked to such core repair systems, including homologous recombination (HR). We revealed that Atxn1 actually interacted with RpA1 and its essential partners BRCA1/2. Furthermore, mutant but not normal Atxn1 impaired the dynamics of RpA1 in the nucleus after DNA damage. Uptake of BrdU by Purkinje cells was observed in mutant Atxn1 knockin mice, suggesting their abnormal entry to the S-phase. In addition, chemical and genetic inhibitions of Chk1 elongated lifespan and recovered eye degeneration. Collectively, we elucidated core networks for DNA damage repair in SCA1 that might include the aberrant usage of HR.


Assuntos
Dano ao DNA , Reparo do DNA , Drosophila/genética , Redes Reguladoras de Genes , Ataxias Espinocerebelares/genética , Animais , Animais Geneticamente Modificados , Ataxina-1 , Ataxinas , Ciclo Celular/genética , Quinase 1 do Ponto de Checagem , Modelos Animais de Doenças , Feminino , Vetores Genéticos/genética , Humanos , Longevidade/genética , Masculino , Mutagênese Insercional , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Proteínas Quinases/metabolismo , Células de Purkinje/metabolismo , Transdução de Sinais , Ataxias Espinocerebelares/metabolismo , Ataxias Espinocerebelares/mortalidade , Biologia de Sistemas
7.
Hum Mol Genet ; 21(5): 1099-110, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22100762

RESUMO

The spinocerebellar ataxia type 7 (SCA7) gene product, Ataxin-7 (ATXN7), localizes to the nucleus and has been shown to function as a component of the TATA-binding protein-free TAF-containing-SPT3-TAF9-GCN5-acetyltransferase transcription complex, although cytoplasmic localization of ATXN7 in affected neurons of human SCA7 patients has also been detected. Here, we define a physiological function for cytoplasmic ATXN7. Live imaging reveals that the intracellular distribution of ATXN7 dynamically changes and that ATXN7 distribution frequently shifts from the nucleus to the cytoplasm. Immunocytochemistry and immunoprecipitation demonstrate that cytoplasmic ATXN7 associates with microtubules (MTs), and expression of ATXN7 stabilizes MTs against nocodazole treatment, while ATXN7 knockdown enhances MT degradation. Interestingly, normal and mutant ATXN7 similarly associate with and equally stabilize MTs. Taken together, these findings provide a novel physiological function of ATXN7 in the regulation of cytoskeletal dynamics, and suggest that abnormal cytoskeletal regulation may contribute to SCA7 disease pathology.


Assuntos
Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Tubulina (Proteína)/metabolismo , Ataxina-7 , Ciclo Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Células HeLa , Humanos , Microtúbulos/ultraestrutura , Mitose , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Domínios e Motivos de Interação entre Proteínas , Interferência de RNA , Transfecção
8.
EMBO J ; 29(14): 2446-60, 2010 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-20531390

RESUMO

Non-cell-autonomous effect of mutant proteins expressed in glia has been implicated in several neurodegenerative disorders, whereas molecules mediating the toxicity are currently not known. We identified a novel molecule named multiple alpha-helix protein located at ER (Maxer) downregulated by mutant ataxin-1 (Atx1) in Bergmann glia. Maxer is an endoplasmic reticulum (ER) membrane protein interacting with CDK5RAP3. Maxer anchors CDK5RAP3 to the ER and inhibits its function of Cyclin D1 transcription repression in the nucleus. The loss of Maxer eventually induces cell accumulation at G1 phase. It was also shown that mutant Atx1 represses Maxer and inhibits proliferation of Bergmann glia in vitro. Consistently, Bergmann glia are reduced in the cerebellum of mutant Atx1 knockin mice before onset. Glutamate-aspartate transporter reduction in Bergmann glia by mutant Atx1 and vulnerability of Purkinje cell to glutamate are both strengthened by Maxer knockdown in Bergmann glia, whereas Maxer overexpression rescues them. Collectively, these results suggest that the reduction of Maxer mediates functional deficiency of Bergmann glia, and might contribute to the non-cell-autonomous pathology of SCA1.


Assuntos
Retículo Endoplasmático/metabolismo , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso , Neuroglia/metabolismo , Proteínas Nucleares , Sequência de Aminoácidos , Animais , Ataxina-1 , Ataxinas , Proteínas de Ciclo Celular , Proliferação de Células , Transportador 1 de Aminoácido Excitatório/genética , Transportador 1 de Aminoácido Excitatório/metabolismo , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Neuroglia/citologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Ratos , Ratos Wistar , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Distribuição Tecidual , Proteínas Supressoras de Tumor
9.
Nat Cell Biol ; 9(4): 402-14, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17384639

RESUMO

Nuclear dysfunction is a key feature of the pathology of polyglutamine (polyQ) diseases. It has been suggested that mutant polyQ proteins impair functions of nuclear factors by interacting with them directly in the nucleus. However, a systematic analysis of quantitative changes in soluble nuclear proteins in neurons expressing mutant polyQ proteins has not been performed. Here, we perform a proteome analysis of soluble nuclear proteins prepared from neurons expressing huntingtin (Htt) or ataxin-1 (AT1) protein, and show that mutant AT1 and Htt similarly reduce the concentration of soluble high mobility group B1/2 (HMGB1/2) proteins. Immunoprecipitation and pulldown assays indicate that HMGBs interact with mutant AT1 and Htt. Immunohistochemistry showed that these proteins were reduced in the nuclear region outside of inclusion bodies in affected neurons. Compensatory expression of HMGBs ameliorated polyQ-induced pathology in primary neurons and in Drosophila polyQ models. Furthermore, HMGBs repressed genotoxic stress signals induced by mutant Htt or transcriptional repression. Thus, HMGBs may be critical regulators of polyQ disease pathology and could be targets for therapy development.


Assuntos
Proteína HMGB1/fisiologia , Proteína HMGB2/fisiologia , Doenças Neurodegenerativas/metabolismo , Proteínas Nucleares/fisiologia , Proteômica/métodos , Animais , Western Blotting , Morte Celular , Células Cultivadas , Drosophila , Eletroforese em Gel Bidimensional , Proteína HMGB1/análise , Proteína HMGB1/metabolismo , Proteína HMGB2/análise , Proteína HMGB2/metabolismo , Imuno-Histoquímica , Imunoprecipitação , Modelos Biológicos , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Neurônios/citologia , Neurônios/metabolismo , Proteínas Nucleares/análise , Proteínas Nucleares/metabolismo , Peptídeos/genética , Peptídeos/metabolismo , Ligação Proteica , Células de Purkinje/citologia , Células de Purkinje/metabolismo , RNA Interferente Pequeno , Ratos , Ratos Wistar , Transdução de Sinais , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
10.
Commun Med (Lond) ; 3(1): 170, 2023 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-38017287

RESUMO

BACKGROUND: Charcot-Marie-Tooth disease type 1A (CMT1A) is one of the most common hereditary peripheral neuropathies caused by duplication of 1.5 Mb genome region including PMP22 gene. We aimed to correct the duplication in human CMT1A patient-derived iPS cells (CMT1A-iPSCs) by genome editing and intended to analyze the effect on Schwann cells differentiated from CMT1A-iPSCs. METHODS: We designed multiple gRNAs targeting a unique sequence present at two sites that sandwich only a single copy of duplicated peripheral myelin protein 22 (PMP22) genes, and selected one of them (gRNA3) from screening their efficiencies by T7E1 mismatch detection assay. AAV2-hSaCas9-gRNAedit was generated by subcloning gRNA3 into pX601-AAV-CMV plasmid, and the genome editing AAV vector was infected to CMT1A-iPSCs or CMT1A-iPSC-derived Schwann cell precursors. The effect of the genome editing AAV vector on myelination was evaluated by co-immunostaining of myelin basic protein (MBP), a marker of mature myelin, and microtubule-associated protein  2(MAP2), a marker of neurites or by electron microscopy. RESULTS: Here we show that infection of CMT1A-iPS cells (iPSCs) with AAV2-hSaCas9-gRNAedit expressing both hSaCas9 and gRNA targeting the tandem repeat sequence decreased PMP22 gene duplication by 20-40%. Infection of CMT1A-iPSC-derived Schwann cell precursors with AAV2-hSaCas9-gRNAedit normalized PMP22 mRNA and PMP22 protein expression levels, and also ameliorated increased apoptosis and impaired myelination in CMT1A-iPSC-derived Schwann cells. CONCLUSIONS: In vivo transfer of AAV2-hSaCas9-gRNAedit to peripheral nerves could be a potential therapeutic modality for CMT1A patient after careful examinations of toxicity including off-target mutations.


Charcot-Marie-Tooth disease type 1A (CMT1A) is a common heritable form of the condition that develops when nerves in the body's extremities, such as the hands, feet and arms, are damaged due to an extra copy of PMP22 gene being incorrectly produced. Currently, no known therapies exist. Here, we developed a method to delete the additional copy of PMP22 gene by 20­40% to prevent overproduction. Our results show that this method can reduce PMP22 protein production, leading to near normal production in patient's nerve cells. Further safety assessments should now be undertaken. If the treatment is safe for patients it could become a therapeutic option for CMT1A patients.

11.
J Cell Biol ; 172(4): 589-604, 2006 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-16461361

RESUMO

Transcriptional disturbance is implicated in the pathology of polyglutamine diseases, including Huntington's disease (HD). However, it is unknown whether transcriptional repression leads to neuronal death or what forms that death might take. We found transcriptional repression-induced atypical death (TRIAD) of neurons to be distinct from apoptosis, necrosis, or autophagy. The progression of TRIAD was extremely slow in comparison with other types of cell death. Gene expression profiling revealed the reduction of full-length yes-associated protein (YAP), a p73 cofactor to promote apoptosis, as specific to TRIAD. Furthermore, novel neuron-specific YAP isoforms (YAPDeltaCs) were sustained during TRIAD to suppress neuronal death in a dominant-negative fashion. YAPDeltaCs and activated p73 were colocalized in the striatal neurons of HD patients and mutant huntingtin (htt) transgenic mice. YAPDeltaCs also markedly attenuated Htt-induced neuronal death in primary neuron and Drosophila melanogaster models. Collectively, transcriptional repression induces a novel prototype of neuronal death associated with the changes of YAP isoforms and p73, which might be relevant to the HD pathology.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Ligação a DNA/metabolismo , Doença de Huntington/metabolismo , Neurônios/patologia , Proteínas Nucleares/metabolismo , Fosfoproteínas/genética , Transcrição Gênica/efeitos dos fármacos , Amanitinas/farmacologia , Sequência de Aminoácidos , Animais , Morte Celular/genética , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Drosophila melanogaster/genética , Pesquisas com Embriões , Genes Supressores de Tumor , Humanos , Doença de Huntington/patologia , Camundongos , Dados de Sequência Molecular , Mutagênese Insercional , Neurônios/metabolismo , Proteínas Nucleares/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Interferente Pequeno/farmacologia , Ratos , Fatores de Tempo , Transativadores/efeitos dos fármacos , Transativadores/fisiologia , Fatores de Transcrição , Proteína Tumoral p73 , Proteínas Supressoras de Tumor , Proteínas de Sinalização YAP
12.
Commun Biol ; 4(1): 961, 2021 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-34385591

RESUMO

Multiple gene mutations cause familial frontotemporal lobar degeneration (FTLD) while no single gene mutations exists in sporadic FTLD. Various proteins aggregate in variable regions of the brain, leading to multiple pathological and clinical prototypes. The heterogeneity of FTLD could be one of the reasons preventing development of disease-modifying therapy. We newly develop a mathematical method to analyze chronological changes of PPI networks with sequential big data from comprehensive phosphoproteome of four FTLD knock-in (KI) mouse models (PGRNR504X-KI, TDP43N267S-KI, VCPT262A-KI and CHMP2BQ165X-KI mice) together with four transgenic mouse models of Alzheimer's disease (AD) and with APPKM670/671NL-KI mice at multiple time points. The new method reveals the common core pathological network across FTLD and AD, which is shared by mouse models and human postmortem brains. Based on the prediction, we performed therapeutic intervention of the FTLD models, and confirmed amelioration of pathologies and symptoms of four FTLD mouse models by interruption of the core molecule HMGB1, verifying the new mathematical method to predict dynamic molecular networks.


Assuntos
Doença de Alzheimer/etiologia , Modelos Animais de Doenças , Degeneração Lobar Frontotemporal/etiologia , Doença de Alzheimer/patologia , Animais , Degeneração Lobar Frontotemporal/patologia , Humanos , Camundongos , Camundongos Transgênicos , Modelos Teóricos
13.
ACS Chem Neurosci ; 12(16): 3015-3027, 2021 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-34319089

RESUMO

Tau aggregation is a central hallmark of tauopathies such as frontotemporal lobar degeneration and progressive supranuclear palsy as well as of Alzheimer's disease, and it has been a target for therapeutic development. Herein, we unexpectedly found that hepta-histidine (7H), an inhibitor of the interaction between Ku70 and Huntingtin proteins, suppresses aggregation of Tau-R3 peptides in vitro. Addition of the trans-activator of transcription (TAT) sequence (YGRKKRRQRRR) derived from the TAT protein to 7H increased its permeability into cells, and TAT-7H treatment of iPS cell-derived neurons carrying Tau or APP mutations suppressed Tau phosphorylation. These results indicate that 7H is a promising lead compound for developing anti-aggregation drugs against Tau-related neurodegenerative diseases including Alzheimer's disease (AD).


Assuntos
Doença de Alzheimer , Degeneração Lobar Frontotemporal , Tauopatias , Doença de Alzheimer/tratamento farmacológico , Histidina , Humanos , Proteínas tau
14.
Commun Biol ; 4(1): 1175, 2021 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-34635772

RESUMO

DNA damage is increased in Alzheimer's disease (AD), while the underlying mechanisms are unknown. Here, we employ comprehensive phosphoproteome analysis, and identify abnormal phosphorylation of 70 kDa subunit of Ku antigen (Ku70) at Ser77/78, which prevents Ku70-DNA interaction, in human AD postmortem brains. The abnormal phosphorylation inhibits accumulation of Ku70 to the foci of DNA double strand break (DSB), impairs DNA damage repair and eventually causes transcriptional repression-induced atypical cell death (TRIAD). Cells under TRIAD necrosis reveal senescence phenotypes. Extracellular high mobility group box 1 (HMGB1) protein, which is released from necrotic or hyper-activated neurons in AD, binds to toll-like receptor 4 (TLR4) of neighboring neurons, and activates protein kinase C alpha (PKCα) that executes Ku70 phosphorylation at Ser77/78. Administration of human monoclonal anti-HMGB1 antibody to post-symptomatic AD model mice decreases neuronal DSBs, suppresses secondary TRIAD necrosis of neurons, prevents escalation of neurodegeneration, and ameliorates cognitive symptoms. TRIAD shares multiple features with senescence. These results discover the HMGB1-Ku70 axis that accounts for the increase of neuronal DNA damage and secondary enhancement of TRIAD, the cell death phenotype of senescence, in AD.


Assuntos
Doença de Alzheimer/patologia , Dano ao DNA , Reparo do DNA , Proteína HMGB1/fisiologia , Autoantígeno Ku/metabolismo , Transdução de Sinais/genética , Animais , Proteína HMGB1/genética , Camundongos , Camundongos Transgênicos , Fosforilação
15.
Life Sci Alliance ; 4(7)2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34130995

RESUMO

The early-stage pathologies of frontotemporal lobal degeneration (FTLD) remain largely unknown. In VCPT262A-KI mice carrying VCP gene mutation linked to FTLD, insufficient DNA damage repair in neural stem/progenitor cells (NSCs) activated DNA-PK and CDK1 that disabled MCM3 essential for the G1/S cell cycle transition. Abnormal neural exit produced neurons carrying over unrepaired DNA damage and induced early-stage transcriptional repression-induced atypical cell death (TRIAD) necrosis accompanied by the specific markers pSer46-MARCKS and YAP. In utero gene therapy expressing normal VCP or non-phosphorylated mutant MCM3 rescued DNA damage, neuronal necrosis, cognitive function, and TDP43 aggregation in adult neurons of VCPT262A-KI mice, whereas similar therapy in adulthood was less effective. The similar early-stage neuronal necrosis was detected in PGRNR504X-KI, CHMP2BQ165X-KI, and TDPN267S-KI mice, and blocked by embryonic treatment with AAV-non-phospho-MCM3. Moreover, YAP-dependent necrosis occurred in neurons of human FTLD patients, and consistently pSer46-MARCKS was increased in cerebrospinal fluid (CSF) and serum of these patients. Collectively, developmental stress followed by early-stage neuronal necrosis is a potential target for therapeutics and one of the earliest general biomarkers for FTLD.


Assuntos
Degeneração Lobar Frontotemporal/patologia , Células-Tronco Neurais/metabolismo , Proteína com Valosina/metabolismo , Animais , Ciclo Celular , Linhagem da Célula/genética , Células Cultivadas , Dano ao DNA/genética , Dano ao DNA/fisiologia , Proteínas de Ligação a DNA/metabolismo , Degeneração Lobar Frontotemporal/líquido cefalorraquidiano , Degeneração Lobar Frontotemporal/genética , Expressão Gênica/genética , Regulação da Expressão Gênica/genética , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Necrose/metabolismo , Necrose/patologia , Células-Tronco Neurais/patologia , Neurônios/metabolismo , Proteína com Valosina/genética
16.
Nat Commun ; 11(1): 507, 2020 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-31980612

RESUMO

The timing and characteristics of neuronal death in Alzheimer's disease (AD) remain largely unknown. Here we examine AD mouse models with an original marker, myristoylated alanine-rich C-kinase substrate phosphorylated at serine 46 (pSer46-MARCKS), and reveal an increase of neuronal necrosis during pre-symptomatic phase and a subsequent decrease during symptomatic phase. Postmortem brains of mild cognitive impairment (MCI) rather than symptomatic AD patients reveal a remarkable increase of necrosis. In vivo imaging reveals instability of endoplasmic reticulum (ER) in mouse AD models and genome-edited human AD iPS cell-derived neurons. The level of nuclear Yes-associated protein (YAP) is remarkably decreased in such neurons under AD pathology due to the sequestration into cytoplasmic amyloid beta (Aß) aggregates, supporting the feature of YAP-dependent necrosis. Suppression of early-stage neuronal death by AAV-YAPdeltaC reduces the later-stage extracellular Aß burden and cognitive impairment, suggesting that preclinical/prodromal YAP-dependent neuronal necrosis represents a target for AD therapeutics.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Proteínas de Ciclo Celular/metabolismo , Fatores de Transcrição/metabolismo , Doença de Alzheimer/líquido cefalorraquidiano , Peptídeos beta-Amiloides/metabolismo , Animais , Núcleo Celular/metabolismo , Disfunção Cognitiva/líquido cefalorraquidiano , Disfunção Cognitiva/patologia , Simulação por Computador , Modelos Animais de Doenças , Retículo Endoplasmático/patologia , Retículo Endoplasmático/ultraestrutura , Feminino , Proteína HMGB1/líquido cefalorraquidiano , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Lisofosfolipídeos/metabolismo , Masculino , Camundongos Transgênicos , Necrose , Neurônios/metabolismo , Neurônios/patologia , Transdução de Sinais , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Imagem com Lapso de Tempo , Proteínas de Sinalização YAP
17.
J Neurosci ; 27(4): 868-80, 2007 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-17251428

RESUMO

The reason why vulnerabilities to mutant polyglutamine (polyQ) proteins are different among neuronal subtypes is mostly unknown. In this study, we compared the gene expression profiles of three types of primary neurons expressing huntingtin (htt) or ataxin-1. We found that heat shock protein 70 (hsp70), a well known chaperone molecule protecting neurons in the polyQ pathology, was dramatically upregulated only by mutant htt and selectively in the granule cells of the cerebellum. Granule cells, which are insensitive to degeneration in the human Huntington's disease (HD) pathology, lost their resistance by suppressing hsp70 with siRNA, whereas cortical neurons, affected in human HD, gained resistance by overexpressing hsp70. This indicates that induction levels of hsp70 are a critical factor for determining vulnerabilities to mutant htt among neuronal subtypes. CAT (chloramphenicol acetyltransferase) assays showed that CBF (CCAAT box binding factor, CCAAT/enhancer binding protein zeta) activated, but p53 repressed transcription of the hsp70 gene in granule cells. Basal and mutant htt-induced expression levels of p53 were remarkably lower in granule cells than in cortical neurons, suggesting that different magnitudes of p53 are linked to distinct induction levels of hsp70. Surprisingly, however, heat shock factor 1 was not activated in granule cells by mutant htt. Collectively, different levels of hsp70 among neuronal subtypes might be involved in selective neuronal death in the HD pathology.


Assuntos
Proteínas de Choque Térmico HSP70/biossíntese , Doença de Huntington/genética , Doença de Huntington/metabolismo , Mutação , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Proteínas Nucleares/genética , Adulto , Idoso , Animais , Gatos , Células Cultivadas , Cerebelo/metabolismo , Cerebelo/patologia , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/fisiologia , Células HeLa , Humanos , Proteína Huntingtina , Doença de Huntington/patologia , Camundongos , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/fisiologia , Neurônios/patologia , Proteínas Nucleares/fisiologia , Ratos , Ratos Wistar
18.
Eur J Neurosci ; 28(1): 30-40, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18662332

RESUMO

Selective vulnerability of neurons is a critical feature of neurodegenerative diseases, but the underlying molecular mechanisms remain largely unknown. We here report that Omi/HtrA2, a mitochondrial protein regulating survival and apoptosis of cells, decreases selectively in striatal neurons that are most vulnerable to the Huntington's disease (HD) pathology. In microarray analysis, Omi/HtrA2 was decreased under the expression of mutant huntingtin (htt) in striatal neurons but not in cortical or cerebellar neurons. Mutant ataxin-1 (Atx-1) did not affect Omi/HtrA2 in any type of neuron. Western blot analysis of primary neurons expressing mutant htt also confirmed the selective reduction of the Omi/HtrA2 protein. Immunohistochemistry with a mutant htt-transgenic mouse line and human HD brains confirmed reduction of Omi/HtrA2 in striatal neurons. Overexpression of Omi/HtrA2 by adenovirus vector reverted mutant htt-induced cell death in primary neurons. These results collectively suggest that the homeostatic but not proapoptotic function of Omi/HtrA2 is linked to selective vulnerability of striatal neurons in HD pathology.


Assuntos
Corpo Estriado/citologia , Doença de Huntington/metabolismo , Proteínas Mitocondriais/metabolismo , Neurônios/metabolismo , Serina Endopeptidases/metabolismo , Animais , Morte Celular , Células Cultivadas , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Serina Peptidase 2 de Requerimento de Alta Temperatura A , Homeostase , Humanos , Proteína Huntingtina , Doença de Huntington/patologia , Camundongos , Camundongos Transgênicos , Análise em Microsséries , Proteínas Mitocondriais/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Wistar , Serina Endopeptidases/genética
19.
eNeuro ; 5(4)2018.
Artigo em Inglês | MEDLINE | ID: mdl-30225354

RESUMO

Phosphorylation of myristoylated alanine-rich C kinase substrate (MARCKS) reflects neurite degeneration at the early stage of Alzheimer's disease (AD), before extracellular Aß aggregates are histologically detectable. Here, we demonstrate that similar changes in MARCKS occur in Parkinson's disease (PD) and dementia with Lewy bodies (DLB) pathologies in both mouse models and human patients. The increase in the level of pSer46-MARCKS began before α-synuclein aggregate formation, at a time when human α-Syn-BAC-Tg/GBA-hetero-KO mice exhibited no symptoms, and was sustained during aging, consistent with the pattern in human postmortem brains. The results strongly imply a common mechanism of pre-aggregation neurite degeneration in AD and PD/DLB pathologies.


Assuntos
Envelhecimento/metabolismo , Doença de Alzheimer , Doença por Corpos de Lewy , Substrato Quinase C Rico em Alanina Miristoilada/metabolismo , Neuritos/patologia , Doença de Parkinson , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Modelos Animais de Doenças , Feminino , Humanos , Doença por Corpos de Lewy/metabolismo , Doença por Corpos de Lewy/patologia , Masculino , Camundongos , Camundongos Transgênicos , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Fosforilação
20.
Nat Commun ; 9(1): 433, 2018 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-29382817

RESUMO

Mutations in the progranulin (PGRN) gene cause a tau pathology-negative and TDP43 pathology-positive form of frontotemporal lobar degeneration (FTLD-TDP). We generated a knock-in mouse harboring the R504X mutation (PGRN-KI). Phosphoproteomic analysis of this model revealed activation of signaling pathways connecting PKC and MAPK to tau prior to TDP43 aggregation and cognitive impairments, and identified PKCα as the kinase responsible for the early-stage tau phosphorylation at Ser203. Disinhibition of Gas6 binding to Tyro3 due to PGRN reduction results in activation of PKCα via PLCγ, inducing tau phosphorylation at Ser203, mislocalization of tau to dendritic spines, and spine loss. Administration of a PKC inhibitor, B-Raf inhibitor, or knockdown of molecules in the Gas6-Tyro3-tau axis rescues spine loss and cognitive impairment of PGRN-KI mice. Collectively, these results suggest that targeting of early-stage and aggregation-independent tau signaling represents a promising therapeutic strategy for this disease.


Assuntos
Degeneração Lobar Frontotemporal/etiologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Proteínas tau/metabolismo , Animais , Modelos Animais de Doenças , Degeneração Lobar Frontotemporal/metabolismo , Técnicas de Introdução de Genes , Granulinas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , Fosfolipase C gama/metabolismo , Fosforilação , Progranulinas , Proteína Quinase C-alfa/metabolismo , Proteínas Proto-Oncogênicas B-raf/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA