Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
EBioMedicine ; 95: 104725, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37517100

RESUMO

BACKGROUND: While advancements in imaging techniques have led to major strides in deciphering the human brain, successful interventions are elusive and represent some of the most persistent translational gaps in medicine. Human restricted CHRFAM7A has been associated with neuropsychiatric disorders. METHODS: The physiological role of CHRFAM7A in human brain is explored using multiomics approach on 600 post mortem human brain tissue samples. The emerging pathways and mechanistic hypotheses are tested and validated in an isogenic hiPSC model of CHRFAM7A knock-in medial ganglionic eminence progenitors and neurons. FINDINGS: CHRFAM7A is identified as a modulator of intracellular calcium dynamics and an upstream regulator of Rac1. Rac1 activation re-designs the actin cytoskeleton leading to dynamic actin driven remodeling of membrane protrusion and a switch from filopodia to lamellipodia. The reinforced cytoskeleton leads to an advantage to tolerate stiffer mechanical properties of the extracellular environment. INTERPRETATION: CHRFAM7A modifies the actin cytoskeleton to a more dynamic and stiffness resistant state in an α7nAChR dependent manner. CHRFAM7A may facilitate neuronal adaptation to changes in the brain environment in physiological and pathological conditions contributing to risk or recovery. Understanding how CHRFAM7A affects human brain requires human studies in the areas of memory formation and erasure, cognitive reserve, and neuronal plasticity. FUNDING: This work is supported in part by the Community Foundation for Greater Buffalo (Kinga Szigeti). Also, in part by the International Society for Neurochemistry (ISN) and The Company of Biologists (Nicolas Rosas). ROSMAP is supported by NIA grants P30AG10161, P30AG72975, R01AG15819, R01AG17917. U01AG46152, and U01AG61356.


Assuntos
Encéfalo , Mutação com Ganho de Função , Humanos , Encéfalo/metabolismo , Neurônios/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo
2.
J Neurochem ; 165(4): 603-621, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36943192

RESUMO

The neuronal membrane glycoprotein M6a (GPM6A) belongs to the family of myelin proteolipid protein and plays a role in neuronal remodeling and plasticity. Decreased expression of GPM6A mRNA is observed in the hippocampal tissue of suicide victims who suffered from depression and after chronic stress exposure in animals. The regulatory mechanisms that impact expression of GPM6A under chronic stress or in pathological conditions are not well understood. Previously, miRNAs miR-133b, miR-124-3p, and miR-9-5p have been shown to regulate the expression of Gpm6a mRNA under normal conditions. Here, we employed the paradigm of chronic-restraint stress in rats and using quantitative polymerase chain reaction (qPCR) showed down-regulation of expression of Gpm6a and the brain-derived neurotrophic factor (Bdnf) genes at mRNA level as well as miR-133b, and miR-124-3p, but not miR-9-5p in the hippocampus of chronically stressed rats. Furthermore, we observed alterations in the expression of histone deacetylase (Hdac5) and myocyte enhancer factor 2C (Mef2c) mRNAs. Our data suggest that chronic stress influences Gpm6a expression by miR-124-mediated impact on the expression of Hdac5 and Mef2c. Upon miR-124 over-expression in hippocampal neurons cultured in vitro, we observed enhanced neuronal arborization as evaluated by Sholl analysis, increased Gpm6a and Mef2c expression, and decreased Hdac5 expression. Moreover, treatment of hippocampal neurons cultured in vitro with BDNF resulted in an elevation in the miR-124-3p expression, a decrease in the miR-9-5p expression but did not affect miR-133b. This was accompanied by augmented expression of Gpm6a and Mef2c mRNAs and significantly lower levels of Hdac5 mRNA. Altogether, these results indicate that the regulatory mechanism that influence expression of Gpm6a under chronic stress involves miR-124-mediated impact on the expression of Hdac5 and Mef2c and a role of BDNF in the activation of Gpm6a expression.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , MicroRNAs , Animais , Ratos , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Regulação para Baixo , Hipocampo/metabolismo , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Neurônios/metabolismo
3.
Front Mol Neurosci ; 11: 314, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30233315

RESUMO

Neuronal membrane glycoprotein M6a (Gpm6a) is a protein with four transmembrane regions and the N- and the C-ends facing the cytosol. It functions in processes of neuronal development, outgrowth of neurites, and formation of filopodia, spines, and synapsis. Molecular mechanisms by which Gpm6a acts in these processes are not fully comprehended. Structural similarities of Gpm6a with tetraspanins led us to hypothesize that, similarly to tetraspanins, the cytoplasmic tails function as connections with cytoskeletal and/or signaling proteins. Here, we demonstrate that the C- but not the N-terminal cytosolic end of Gpm6a is required for the formation of filopodia by Gpm6a in cultured neurons from rat hippocampus and in neuroblastoma cells N2a. Further immunofluorescence microcopy and flow cytometry analysis show that deletion of neither the N- nor the C-terminal intracellular domains interferes with the recognition of Gpm6a by the function-blocking antibody directed against the extracellular part of Gpm6a. Expression levels of both truncation mutants were not affected but we observed decrease in the amount of both truncated proteins on cell surface suggesting that the incapacity of the Gpm6a lacking C-terminus to induce filopodium formation is not due to the lower amount of Gpm6a on cell surface. Following colocalization assays shows that deletion of the C- but not the N-terminus diminishes the association of Gpm6a with clathrin implying involvement of clathrin-mediated trafficking events. Next, using comprehensive alanine scanning mutagenesis of the C-terminus we identify K250, K255, and E258 as the key residues for the formation of filopodia by Gpm6a. Substitution of these charged residues with alanine also diminishes the amount of Gpm6a on cell surface and in case of K255 and E258 leads to the lower amount of total expressed protein. Subsequent bioinformatic analysis of Gpm6a amino acid sequence reveals that highly conserved and functional residues cluster preferentially within the C- and not within the N-terminus and that K250, K255, and E258 are predicted as part of sorting signals of transmembrane proteins. Altogether, our results provide evidence that filopodium outgrowth induced by Gpm6a requires functionally critical residues within the C-terminal cytoplasmic tail.

4.
Neuroscience ; 333: 331-44, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27474226

RESUMO

The p21-activated kinases (PAKs) of group I are the main effectors for the small Rho GTPases, critically involved in neurodevelopment, plasticity and maturation of the nervous system. Moreover, the neuronal complexity controlled by PAK1/PAK3 signaling determines the postnatal brain size and synaptic properties. Stress induces alterations at the level of structural and functional synaptic plasticity accompanied by reductions in size and activity of the hippocampus and the prefrontal cortex (PFC). These abnormalities are likely to contribute to the pathology of depression and, in part, reflect impaired cytoskeleton remodeling pointing to the role of Rho GTPase signaling. Thus, the present study assessed the expression of the group I PAKs and their activators in the brain of depressed subjects. Using quantitative polymerase chain reaction (qPCR), mRNA levels and coexpression of the group I PAKs: PAK1, PAK2, and PAK3 as well as of their activators: RAC1, CDC42 and ARHGEF7 were examined in postmortem samples from the PFC (n=25) and the hippocampus (n=23) of subjects with depression and compared to control subjects (PFC n=24; hippocampus n=21). Results demonstrated that mRNA levels of PAK1 and PAK3, are significantly reduced in the brain of depressed subjects, with PAK1 being reduced in the PFC and PAK3 in the hippocampus. No differences were observed for the ubiquitously expressed PAK2. Following analysis of gene coexpression demonstrated disruption of coordinated gene expression in the brain of subjects with depression. Abnormalities in mRNA expression of PAK1 and PAK3 as well as their altered coexpression patterns were detected in the brain of subjects with depression.


Assuntos
Encéfalo/enzimologia , Transtorno Depressivo/enzimologia , Quinases Ativadas por p21/metabolismo , Adulto , Idoso , Encéfalo/patologia , Estudos de Coortes , Transtorno Depressivo/patologia , Feminino , Proteínas Ativadoras de GTPase/metabolismo , Perfilação da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Fosfoproteínas/metabolismo , Reação em Cadeia da Polimerase , RNA Mensageiro/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Caracteres Sexuais , Adulto Jovem , Proteínas rac1 de Ligação ao GTP/metabolismo
5.
J Neurochem ; 137(1): 46-61, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26809475

RESUMO

Stress-responsive neuronal membrane glycoprotein M6a (Gpm6a) functions in neurite extension, filopodium and spine formation and synaptogenesis. The mechanisms of Gpm6a action in these processes are incompletely understood. Previously, we identified the actin regulator coronin-1a (Coro1a) as a putative Gpm6a interacting partner. Here, we used co-immunoprecipitation assays with the anti-Coro1a antibody to show that Coro1a associates with Gpm6a in rat hippocampal neurons. By immunofluorescence microscopy, we demonstrated that in hippocampal neurons Coro1a localizes in F-actin-enriched regions and some of Coro1a spots co-localize with Gpm6a labeling. Notably, the over-expression of a dominant-negative form of Coro1a as well as its down-regulation by siRNA interfered with Gpm6a-induced filopodium formation. Coro1a is known to regulate the plasma membrane translocation and activation of small GTPase Rac1. We show that Coro1a co-immunoprecipitates with Rac1 together with Gpm6a. Pharmacological inhibition of Rac1 resulted in a significant decrease in filopodium formation by Gpm6a. The same was observed upon the co-expression of Gpm6a with the inactive GDP-bound form of Rac1. In this case, the elevated membrane recruitment of GDP-bound Rac1 was detected as well. Moreover, the kinase activity of the p21-activated kinase 1 (Pak1), a main downstream effector of Rac1 that acts downstream of Coro1a, was required for Gpm6a-induced filopodium formation. Taken together, our results provide evidence that a signaling pathway including Coro1a, Rac1, and Pak1 facilitates Gpm6a-induced filopodium formation. Formation of filopodia by membrane glycoprotein M6a (Gpm6a) requires actin regulator coronin-1a (Coro1a), known to regulate plasma membrane localization and activation of Rac1 and its downstream effector Pak1. Coro1a associates with Gpm6a. Blockage of Coro1a, Rac1, or Pak1 interferes with Gpm6a-induced filopodium formation. Moreover, Gpm6a facilitates Rac1 membrane recruitment. Altogether, a mechanistic insight into the process of Gpm6a-induced neuronal filopodium formation is provided.


Assuntos
Glicoproteínas de Membrana/fisiologia , Proteínas dos Microfilamentos/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neurônios/ultraestrutura , Pseudópodes/fisiologia , Quinases Ativadas por p21/fisiologia , Proteínas rac1 de Ligação ao GTP/fisiologia , Actinas/análise , Animais , Células Cultivadas , Regulação para Baixo , Genes Reporter , Hipocampo/citologia , Proteínas dos Microfilamentos/genética , Proteínas do Tecido Nervoso/antagonistas & inibidores , Biogênese de Organelas , Cultura Primária de Células , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Proteínas rac1 de Ligação ao GTP/antagonistas & inibidores
6.
Exp Cell Res ; 338(2): 170-82, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26226218

RESUMO

Adenovirus serotypes have been shown to cause drastic changes in nuclear organization, including the transcription machinery, during infection. This ability of adenovirus to subvert transcription in the host cell facilitates viral replication. Because nuclear actin and nuclear myosin I, myosin V and myosin VI have been implicated as direct regulators of transcription and important factors in the replication of other viruses, we sought to determine how nuclear actin and myosins are involved in adenovirus infection. We first confirmed reorganization of the host's transcription machinery to viral replication centers. We found that nuclear actin also reorganizes to sites of transcription through the intermediate but not the advanced late phase of viral infection. Furthermore, nuclear myosin I localized with nuclear actin and sites of transcription in viral replication centers. Intriguingly, nuclear myosins V and VI, which also reorganized to viral replication centers, exhibited different localization patterns, suggesting specialized roles for these nuclear myosins. Finally, we assessed the role of actin in adenovirus infection and found both cytoplasmic and nuclear actin likely play roles in adenovirus infection and replication. Together our data suggest the involvement of actin and multiple myosins in the nuclear replication and late viral gene expression of adenovirus.


Assuntos
Actinas/metabolismo , Infecções por Adenoviridae/metabolismo , Núcleo Celular/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Miosina Tipo V/metabolismo , Adenovírus Humanos/genética , Linhagem Celular Tumoral , Núcleo Celular/virologia , Citoplasma/metabolismo , Citoplasma/virologia , Replicação do DNA/genética , Células HeLa , Humanos , Transcrição Gênica/fisiologia , Replicação Viral/genética
7.
J Biol Chem ; 284(46): 32075-88, 2009 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-19737934

RESUMO

Gpm6a was identified as a stress-responsive gene in the hippocampal formation. This gene is down-regulated in the hippocampus of both socially and physically stressed animals, and this effect can be reversed by antidepressant treatment. Previously we showed that the stress-regulated protein M6a is a key modulator for neurite outgrowth and filopodium/spine formation. In the present work, mutational analysis was used to characterize the action of M6a at the molecular level. We show that four cysteines 162, 174, 192, and 202 within EC2 are functionally crucial sites. The presence of cysteines 162 and 202 is essential for the efficient cell surface expression of the M6a protein. In contrast, cysteines 174 and 192, which form a disulfide bridge as shown by biochemical analysis, are not required for the efficient surface expression of M6a. Their mutation to alanine does not interfere with the localization of M6a to filopodial protrusions in primary hippocampal neurons. The neurons expressing C174A and/or C192A mutants display decreased filopodia number. In non-permeabilized cells, these mutant proteins are not recognized by a function-blocking monoclonal antibody directed to M6a. Moreover, neurons in contact with axons expressing C174A/C192A mutant display significantly lower density of presynaptic clusters over their dendrites. Taken together, this study demonstrates that cysteines in the EC2 domain are critical for the role of M6a in filopodium outgrowth and synaptogenesis.


Assuntos
Membrana Celular/metabolismo , Cisteína/genética , Hipocampo/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Estresse Fisiológico , Animais , Western Blotting , Células COS , Células Cultivadas , Chlorocebus aethiops , Cisteína/metabolismo , Hipocampo/citologia , Técnicas Imunoenzimáticas , Glicoproteínas de Membrana/genética , Mutagênese Sítio-Dirigida , Proteínas do Tecido Nervoso/genética , Ratos , Ratos Wistar/embriologia
8.
Curr Biol ; 16(8): 825-31, 2006 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-16631592

RESUMO

Increasing evidence suggests functional compartmentalization of interphase nuclei. This includes preferential interior localization of gene-rich and early replicating chromosome regions versus peripheral localization of gene-poor and late replicating chromosome regions , association of some active genes with nuclear speckles or transcription "factories", and association of transcriptionally repressed genes with heterochromatic regions. Dynamic changes in chromosome compartmentalization imply mechanisms for long-range interphase chromatin movements. However, live cell imaging in mammalian cells has revealed limited chromatin mobility, described as "constrained diffusion". None of these studies, though, have examined a chromosome locus undergoing an inducible repositioning between two different nuclear compartments. Here we demonstrate migration of an interphase chromosome site from the nuclear periphery to the interior 1-2 hr after targeting a transcriptional activator to this site. Spot redistribution is perturbed by specific actin or nuclear myosin I mutants. Extended periods of chromosome immobility are interspersed with several minute periods in which chromosomes move unidirectionally along curvilinear paths oriented roughly perpendicular to the nuclear envelope at velocities of 0.1-0.9 microm/min over distances of 1-5 microm. Our results suggest an active mechanism for fast and directed long-range interphase chromosome movements dependent directly or indirectly on actin/myosin.


Assuntos
Posicionamento Cromossômico/fisiologia , Interfase/fisiologia , Actinas/fisiologia , Animais , Células CHO/citologia , Cromossomos/metabolismo , Cricetinae , Cricetulus , Movimento/fisiologia , Miosinas/fisiologia , Fatores de Tempo
9.
Nat Cell Biol ; 6(11): 1094-101, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15502823

RESUMO

Actin is abundant in the nucleus and has been implicated in transcription; however, the nature of this involvement has not been established. Here we demonstrate that beta-actin is critically involved in transcription because antibodies directed against beta-actin, but not muscle actin, inhibited transcription in vivo and in vitro. Chromatin immunoprecipitation assays demonstrated the recruitment of actin to the promoter region of the interferon-gamma-inducible MHC2TA gene as well as the interferon-alpha-inducible G1P3 gene. Further investigation revealed that actin and RNA polymerase II co-localize in vivo and also co-purify. We employed an in vitro system with purified nuclear components to demonstrate that antibodies to beta-actin block the initiation of transcription. This assay also demonstrates that beta-actin stimulates transcription by RNA polymerase II. Finally, DNA-binding experiments established the presence of beta-actin in pre-initiation complexes and also showed that the depletion of actin prevented the formation of pre-initiation complexes. Together, these data suggest a fundamental role for actin in the initiation of transcription by RNA polymerase II.


Assuntos
Actinas/fisiologia , RNA Polimerase II/fisiologia , Transcrição Gênica/fisiologia , Sequência de Bases , Primers do DNA , Células HeLa , Humanos , Proteínas Mitocondriais , Proteínas Nucleares/genética , Regiões Promotoras Genéticas , Proteínas/genética , Transativadores/genética
10.
J Cell Biol ; 158(3): 463-73, 2002 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-12163469

RESUMO

It is known that nuclear DNA helicase II (NDH II) links CREB-binding protein directly to RNA polymerase II holoenzyme, and that this interaction is essential for gene activation by CREB. Here, we report for the first time that some NDH II/RNA helicase A is a component of promyelocytic leukemia nuclear bodies (PML NBs). An autoimmune serum specific for PML NBs was identified and used in immunoprecipitation experiments. NDH II was present in the immunoprecipitates as shown by mass spectrometry and by immunoblotting. Immunofluorescence and ultrastructural studies showed that NDH II colocalizes with a small subset of PML NBs in control cells, however, colocalizes with practically all bodies in interferon-alpha-stimulated cells. After interferon stimulation, more PML NBs were found to contain newly synthesized RNA, as indicated by bromouridine incorporation. PML NBs also contain RNA polymerase II. The association of NDH II with PML NBs was transcriptionally dependent, and NDH II was present in all bodies with nascent RNA. Blocking of mRNA synthesis caused NDH II relocalization from nucleoplasm to nucleoli. Based on the data, we suggest that NDH II recruitment to PML NBs is connected with transcriptional regulation of interferon-alpha-inducible genes attached to PML NBs.


Assuntos
Adenosina Trifosfatases/metabolismo , Estruturas do Núcleo Celular/enzimologia , DNA Helicases/metabolismo , Células Eucarióticas/enzimologia , Interferon-alfa/genética , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares , RNA Mensageiro/biossíntese , Fatores de Transcrição/metabolismo , Transcrição Gênica/genética , Adenosina Trifosfatases/genética , Amanitinas/farmacologia , Doenças Autoimunes/imunologia , Proteínas Sanguíneas/imunologia , Proteínas Sanguíneas/farmacologia , Estruturas do Núcleo Celular/ultraestrutura , DNA/biossíntese , DNA/genética , DNA Helicases/genética , Células Eucarióticas/ultraestrutura , Células HeLa , Humanos , Imuno-Histoquímica , Interferon-alfa/farmacologia , Substâncias Macromoleculares , Microscopia Eletrônica , Proteínas de Neoplasias/genética , Inibidores da Síntese de Ácido Nucleico/farmacologia , Testes de Precipitina , Proteína da Leucemia Promielocítica , RNA Mensageiro/genética , Fatores de Transcrição/genética , Transcrição Gênica/efeitos dos fármacos , Proteínas Supressoras de Tumor
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...