Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
J Neurochem ; 159(1): 29-60, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33638179

RESUMO

MECP2 duplication syndrome (MDS), a rare X-linked genomic disorder affecting predominantly males, is caused by duplication of the chromosomal region containing the methyl CpG binding protein-2 (MECP2) gene, which encodes methyl-CpG-binding protein 2 (MECP2), a multi-functional protein required for proper brain development and maintenance of brain function during adulthood. Disease symptoms include severe motor and cognitive impairment, delayed or absent speech development, autistic features, seizures, ataxia, recurrent respiratory infections, and shortened lifespan. The cellular and molecular mechanisms by which a relatively modest increase in MECP2 protein causes such severe disease symptoms are poorly understood and consequently there are no treatments available for this fatal disorder. This review summarizes what is known to date about the structure and zcomplex regulation of MECP2 and its many functions in the developing and adult brain. Additionally, recent experimental findings on the cellular and molecular underpinnings of MDS based on cell culture and mouse models of the disorder are reviewed. The emerging picture from these studies is that MDS is a neurodegenerative disorder in which neurons die in specific parts of the central nervous system, including the cortex, hippocampus, cerebellum, and spinal cord. Neuronal death likely results from astrocytic dysfunction, including a breakdown of glutamate homeostatic mechanisms. The role of elevations in the expression of glial acidic fibrillary protein (GFAP) in astrocytes and the microtubule-associated protein, Tau, in neurons to the pathogenesis of MDS is discussed. Lastly, potential therapeutic strategies to potentially treat MDS are discussed.


Assuntos
Encéfalo/metabolismo , Deficiência Intelectual Ligada ao Cromossomo X/genética , Deficiência Intelectual Ligada ao Cromossomo X/metabolismo , Proteína 2 de Ligação a Metil-CpG/genética , Proteína 2 de Ligação a Metil-CpG/metabolismo , Animais , Encéfalo/patologia , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Humanos , Deficiência Intelectual Ligada ao Cromossomo X/patologia , Mutação/fisiologia
2.
Int J Mol Sci ; 22(11)2021 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-34072862

RESUMO

Alzheimer's disease (AD) is a mostly sporadic brain disorder characterized by cognitive decline resulting from selective neurodegeneration in the hippocampus and cerebral cortex whereas Huntington's disease (HD) is a monogenic inherited disorder characterized by motor abnormalities and psychiatric disturbances resulting from selective neurodegeneration in the striatum. Although there have been numerous clinical trials for these diseases, they have been unsuccessful. Research conducted over the past three decades by a large number of laboratories has demonstrated that abnormal actions of common kinases play a key role in the pathogenesis of both AD and HD as well as several other neurodegenerative diseases. Prominent among these kinases are glycogen synthase kinase (GSK3), p38 mitogen-activated protein kinase (MAPK) and some of the cyclin-dependent kinases (CDKs). After a brief summary of the molecular and cell biology of AD and HD this review covers what is known about the role of these three groups of kinases in the brain and in the pathogenesis of the two neurodegenerative disorders. The potential of targeting GSK3, p38 MAPK and CDKS as effective therapeutics is also discussed as is a brief discussion on the utilization of recently developed drugs that simultaneously target two or all three of these groups of kinases. Multi-kinase inhibitors either by themselves or in combination with strategies currently being used such as immunotherapy or secretase inhibitors for AD and knockdown for HD could represent a more effective therapeutic approach for these fatal neurodegenerative diseases.


Assuntos
Doença de Alzheimer/genética , Glicogênio Sintase Quinase 3 beta/genética , Quinase 3 da Glicogênio Sintase/genética , Doença de Huntington/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Doença de Alzheimer/terapia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Humanos , Doença de Huntington/terapia , Terapia de Alvo Molecular
3.
Dev Neurosci ; 41(3-4): 149-165, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31982872

RESUMO

Neurodevelopment is a highly complex process composed of several carefully regulated events starting from the proliferation of neuroepithelial cells and culminating with and refining of neural networks and synaptic transmission. Improper regulation of any of these neurodevelopmental events often results in severe brain dysfunction. Accumulating evidence indicates that epigenetic modifications of chromatin play a key role in neurodevelopmental regulation. Among these modifications are histone acetylation and deacetylation, which control access of transcription factors to DNA, thereby regulating gene transcription. Histone deacetylation, which restricts access of transcription factor repressing gene transcription, involves the action of members of a family of 18 enzymes, the histone deacetylases (HDAC), which are subdivided in 4 subgroups. This review focuses on the Group 1 HDACs - HDAC 1, 2, 3, and 8. Although much of the evidence for HDAC involvement in neurodevelopment has come from the use of pharmacological inhibitors, because these agents are generally nonselective with regard to their effects on individual members of the HDAC family, this review is limited to evidence garnered from the use of molecular genetic approaches. Our review describes that Class I HDACs play essential roles in all phases of neurodevelopment. Modulation of the activity of individual HDACs could be an important therapeutic approach for neurodevelopmental and psychiatric disorders.


Assuntos
Encéfalo/crescimento & desenvolvimento , Histona Desacetilases/metabolismo , Transtornos Mentais/metabolismo , Fatores de Transcrição/metabolismo , Acetilação , Animais , Encéfalo/metabolismo , Epigênese Genética/genética , Humanos , Transtornos Mentais/fisiopatologia
4.
BMC Neurosci ; 20(1): 65, 2019 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-31883511

RESUMO

BACKGROUND: Histone deacetylase-3 (HDAC3) promotes neurodegeneration in various cell culture and in vivo models of neurodegeneration but the mechanism by which HDAC3 exerts neurotoxicity is not known. HDAC3 is known to be a transcriptional co-repressor. The goal of this study was to identify transcriptional targets of HDAC3 in an attempt to understand how it promotes neurodegeneration. RESULTS: We used chromatin immunoprecipitation analysis coupled with deep sequencing (ChIP-Seq) to identify potential targets of HDAC3 in cerebellar granule neurons. One of the genes identified was the activity-dependent and neuroprotective transcription factor, Neuronal PAS Domain Protein 4 (Npas4). We confirmed using ChIP that in healthy neurons HDAC3 associates weakly with the Npas4 promoter, however, this association is robustly increased in neurons primed to die. We find that HDAC3 also associates differentially with the brain-derived neurotrophic factor (Bdnf) gene promoter, with higher association in dying neurons. In contrast, association of HDAC3 with the promoters of other neuroprotective genes, including those encoding c-Fos, FoxP1 and Stat3, was barely detectable in both healthy and dying neurons. Overexpression of HDAC3 leads to a suppression of Npas4 and Bdnf expression in cortical neurons and treatment with RGFP966, a chemical inhibitor of HDAC3, resulted in upregulation of their expression. Expression of HDAC3 also repressed Npas4 and Bdnf promoter activity. CONCLUSION: Our results suggest that Bdnf and Npas4 are transcriptional targets of Hdac3-mediated repression. HDAC3 inhibitors have been shown to protect against behavioral deficits and neuronal loss in mouse models of neurodegeneration and it is possible that these inhibitors work by upregulating neuroprotective genes like Bdnf and Npas4.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Cerebelo/metabolismo , Histona Desacetilases/metabolismo , Neurônios/metabolismo , Acrilamidas/farmacologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fator Neurotrófico Derivado do Encéfalo/genética , Células Cultivadas , Cerebelo/efeitos dos fármacos , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica/fisiologia , Inibidores de Histona Desacetilases/farmacologia , Neurônios/efeitos dos fármacos , Fenilenodiaminas/farmacologia , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos Wistar , Proteínas Repressoras/metabolismo , Fator de Transcrição STAT3/metabolismo , Transcrição Gênica/fisiologia
5.
J Neurosci ; 37(27): 6575-6587, 2017 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-28550168

RESUMO

Huntington's disease (HD) is an inherited neurodegenerative disease caused by a polyglutamine expansion in the huntington protein (htt). The neuropathological hallmark of HD is the loss of neurons in the striatum and, to a lesser extent, in the cortex. Foxp1 is a member of the Forkhead family of transcription factors expressed selectively in the striatum and the cortex. In the brain, three major Foxp1 isoforms are expressed: isoform-A (∼90 kDa), isoform-D (∼70 kDa), and isoform-C (∼50 kDa). We find that expression of Foxp1 isoform-A and -D is selectively reduced in the striatum and cortex of R6/2 HD mice as well as in the striatum of HD patients. Furthermore, expression of mutant htt in neurons results in the downregulation of Foxp1 Elevating expression of isoform-A or -D protects cortical neurons from death caused by the expression of mutant htt On the other hand, knockdown of Foxp1 promotes death in otherwise healthy neurons. Neuroprotection by Foxp1 is likely to be mediated by the transcriptional stimulation of the cell-cycle inhibitory protein p21Waf1/Cip1 Consistently, Foxp1 activates transcription of the p21Waf1/Cip1 gene promoter, and overexpression of Foxp1 in neurons results in the elevation of p21 expression. Moreover, knocking down of p21Waf1/Cip1 blocks the ability of Foxp1 to protect neurons from mut-Htt-induced neurotoxicity. We propose that the selective vulnerability of neurons of the striatum and cortex in HD is related to the loss of expression of Foxp1, a protein that is highly expressed in these neurons and required for their survival.SIGNIFICANCE STATEMENT Although the mutant huntingtin gene is expressed widely, neurons of the striatum and cortex are selectively affected in Huntington's disease (HD). Our results suggest that this selectivity is attributable to the reduced expression of Foxp1, a protein expressed selectively in striatal and cortical neurons that plays a neuroprotective role in these cells. We show that protection by Foxp1 involves stimulation of the p21Waf1/Cip1 (Cdkn1a) gene. Although three major Foxp1 isoforms (A, C, and D) are expressed in the brain, only isoform-A has been studied in the nervous system. We show that isoform-D is also expressed selectively, neuroprotective and downregulated in HD mice and patients. Our results suggest that Foxp1 might be an attractive therapeutic target for HD.


Assuntos
Córtex Cerebral/metabolismo , Corpo Estriado/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Proteína Huntingtina/metabolismo , Doença de Huntington/metabolismo , Neurônios/metabolismo , Proteínas Repressoras/metabolismo , Animais , Biomarcadores/metabolismo , Células Cultivadas , Córtex Cerebral/patologia , Corpo Estriado/patologia , Regulação para Baixo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Neurônios/patologia , Distribuição Tecidual
6.
J Neurochem ; 145(2): 96-110, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29355955

RESUMO

By their ability to shatter quality of life for both patients and caregivers, neurodegenerative diseases are the most devastating of human disorders. Unfortunately, there are no effective or long-terms treatments capable of slowing down the relentless loss of neurons in any of these diseases. One impediment is the lack of detailed knowledge of the molecular mechanisms underlying the processes of neurodegeneration. While some neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis, are mostly sporadic in nature, driven by both environment and genetic susceptibility, many others, including Huntington's disease, spinocerebellar ataxias, and spinal-bulbar muscular atrophy, are genetically inherited disorders. Surprisingly, given their different roots and etiologies, both sporadic and genetic neurodegenerative disorders have been linked to disease mechanisms involving histone deacetylase (HDAC) proteins, which consists of 18 family members with diverse functions. While most studies have implicated certain HDAC subtypes in promoting neurodegeneration, a substantial body of literature suggests that other HDAC proteins can preserve neuronal viability. Of particular interest, however, is the recent realization that a single HDAC subtype can have both neuroprotective and neurotoxic effects. Diverse mechanisms, beyond transcriptional regulation have been linked to these effects, including deacetylation of non-histone proteins, protein-protein interactions, post-translational modifications of the HDAC proteins themselves and direct interactions with disease proteins. The roles of these HDACs in both sporadic and genetic neurodegenerative diseases will be discussed in the current review.


Assuntos
Histona Desacetilases/metabolismo , Doenças Neurodegenerativas/enzimologia , Animais , Humanos
7.
J Biol Chem ; 291(39): 20787-97, 2016 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-27510036

RESUMO

NPM1 (nucleophosmin 1) is a nucleolar phosphoprotein that regulates many cellular processes, including ribosome biogenesis, proliferation, and genomic integrity. Although its role in proliferating cell types and tissues has been extensively investigated, little is known about its function in neurons and in the brain where it is highly expressed. We report that NPM1 protein expression is increased selectively in the striatum in both the R6/2 transgenic and 3-nitropropionic acid-injected mouse models of Huntington's disease. Examination of the effect of ectopic expression on cultured neurons revealed that increasing NPM1 is toxic to otherwise healthy cerebellar granule and cortical neurons. Toxicity is dependent on its cytoplasmic localization and oligomerization status. Forced retention of NPM1 in the nucleus, as well as inhibiting its ability to oligomerize, not only neutralizes NPM1 toxicity but also renders it protective against apoptosis. Although not blocked by pharmacological inhibition of the pro-apoptotic molecules, JNK, glycogen synthase kinase 3 beta, or caspases, toxicity is blocked by compounds targeting cyclin-dependent kinases (CDKs), as well as by dominant-negative forms of CDK1 and CDK2 and the pan-CDK inhibitor, p21(Cip1/Waf1) Although induced in in vivo Huntington's disease models, NPM1 protein levels are unchanged in cultured cerebellar granule and cortical neurons induced to die by low potassium or homocysteic acid treatment, respectively. Moreover, and counterintuitively, knockdown of its expression or inhibition of endogenous NPM1 oligomerization in these cultured neurons is toxic. Taken together, our study suggests that although neurons need NPM1 for survival, an increase in its expression beyond physiological levels and its translocation to the cytoplasm leads to death through abortive cell cycle induction.


Assuntos
Ciclo Celular , Corpo Estriado/metabolismo , Doença de Huntington/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/biossíntese , Multimerização Proteica , Animais , Proteína Quinase CDC2 , Células Cultivadas , Corpo Estriado/patologia , Quinase 2 Dependente de Ciclina/genética , Quinase 2 Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Quinases Ciclina-Dependentes/genética , Quinases Ciclina-Dependentes/metabolismo , Modelos Animais de Doenças , Doença de Huntington/genética , Doença de Huntington/patologia , Camundongos , Camundongos Transgênicos , Neurônios/patologia , Proteínas Nucleares/genética , Nucleofosmina , Ratos , Ratos Wistar
8.
J Neurosci ; 34(5): 1599-612, 2014 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-24478344

RESUMO

Heat shock factor 1 (HSF1) protects neurons from death caused by the accumulation of misfolded proteins. It is believed that this protective effect is mediated by the transcriptional stimulation of genes encoding heat shock proteins (HSPs), a family of chaperones that refold or degrade misfolded proteins. Whether HSF1 is protective when neuronal death is not caused by protein misfolding has not been studied. Here, we report that HSF1 expression is necessary for the survival of rat neurons and that HSF1 mRNA and protein expression is reduced in neurons primed to die. Knock-down of HSF1 induces death of otherwise healthy neurons, whereas reestablishment of elevated levels of HSF1 protects neurons even when death is not due to accumulation of misfolded proteins. Neuroprotection by HSF1 does not require its trimerization, an event obligatory for the binding of HSF1 to heat shock elements within HSP gene promoters. Moreover, knock-down of HSP70 or blockade of HSP90 signaling does not reduce neuroprotection by HSF1. Although several neuroprotective molecules and signaling pathways, including CaMK, PKA, Casein kinase-II, and the Raf-MEK-ERK and PI-3K-Akt pathways, are not required for HSF1-mediated neuroprotection, protection is abrogated by inhibition of classical histone deacetylases (HDACs). We report that the novel mechanism of neuroprotection by HSF1 involves cooperation with SIRT1, an HDAC with well documented neuroprotective effects. Using a cell culture model of Huntington's disease, we show that HSF1 trimerization is not required for protection against mutant huntingtin-induced neurotoxicity, suggesting that HSF1 can protect neurons against both proteinopathic and nonproteinopathic death through a noncanonical pathway.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Choque Térmico/metabolismo , Neurônios/metabolismo , Fármacos Neuroprotetores/metabolismo , Multimerização Proteica/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Apoptose/genética , Encéfalo/citologia , Células Cultivadas , Imunoprecipitação da Cromatina , Proteínas de Ligação a DNA/genética , Inibidores Enzimáticos/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Fatores de Transcrição de Choque Térmico , Proteínas de Choque Térmico/genética , Humanos , Masculino , Camundongos , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Niacinamida/farmacologia , Estrutura Terciária de Proteína/efeitos dos fármacos , Estrutura Terciária de Proteína/genética , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fatores de Tempo , Fatores de Transcrição/genética , Complexo Vitamínico B/farmacologia
9.
J Biol Chem ; 289(51): 35409-20, 2014 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-25331946

RESUMO

SIRT1, a class III histone deacetylase, protects neurons in various models of neurodegenerative diseases. We previously described that neuroprotection by SIRT1 is independent of its catalytic activity. To elucidate how SIRT1 protects neurons, we performed a mass spectrometric screen to find SIRT1-interacting proteins. One of the proteins identified was JAZ (Znf346), a member of a new class of Cys-2-His-2 zinc finger proteins. To investigate the significance of JAZ in the regulation of neuronal survival, we overexpressed it in neurons. We found that JAZ protects cerebellar granule neurons against potassium deprivation-induced death and cortical neurons from death resulting from oxidative stress. JAZ also protects neurons against toxicity induced by mutant huntingtin and mutant ataxin-1 expression. Although expression of endogenous JAZ does not change in neurons primed to die, knockdown of its expression promotes death of otherwise healthy neurons. In contrast to its protective effect in neurons, overexpression of JAZ in different cell lines promotes death. We find that JAZ suppresses cell cycle progression, thereby explaining its contrasting effect in postmitotic neurons versus proliferating cell lines. Although not affecting the expression of several cyclins, overexpression of JAZ stimulates expression of p21 (WAF1/CIP1), a cell cycle inhibitor known to have neuroprotective effects. Results of chromatin immunoprecipitation and transcriptional assays indicate that the stimulatory effect of JAZ on p21 expression is mediated at the transcriptional level. Furthermore, knockdown of p21 expression inhibits the neuroprotective effect of JAZ. Together, our results suggest that JAZ protects neurons by inhibiting cell cycle re-entry through the transcriptional stimulation of p21 expression.


Assuntos
Proteínas de Transporte/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Sirtuína 1/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Western Blotting , Proteínas de Transporte/genética , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Cultivadas , Cerebelo/citologia , Córtex Cerebral/citologia , Inibidor de Quinase Dependente de Ciclina p21/genética , Expressão Gênica , Células HEK293 , Humanos , Microscopia de Fluorescência , Neurônios/citologia , Neurônios/efeitos dos fármacos , Proteínas Nucleares/genética , Potássio/metabolismo , Potássio/farmacologia , Ligação Proteica , Interferência de RNA , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sirtuína 1/genética
10.
J Biol Chem ; 289(50): 34569-82, 2014 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-25339172

RESUMO

The functional role of histone deacetylase 3 (HDAC3) in the developing brain has yet to be elucidated. We show that mice lacking HDAC3 in neurons and glia of the central nervous system, Nes-Cre/HDAC3 conditional KO mice, show major abnormalities in the cytoarchitecture of the neocortex and cerebellum and die within 24 h of birth. Later-born neurons do not localize properly in the cortex. A similar mislocalization is observed with cerebellar Purkinje neurons. Although the proportion of astrocytes is higher than normal, the numbers of oligodendrocytes are reduced. In contrast, conditional knockout of HDAC3 in neurons of the forebrain and certain other brain regions, using Thy1-Cre and calcium/calmodulin dependent protein kinase II α-Cre for ablation, produces no overt abnormalities in the organization of cells within the cortex or of cerebellar Purkinje neurons at birth. However, both lines of conditional knockout mice suffer from progressive hind limb paralysis and ataxia and die around 6 weeks after birth. The mice display an increase in overall numbers of cells, higher numbers of astrocytes, and Purkinje neuron degeneration. Taken together, our results demonstrate that HDAC3 plays an essential role in regulating brain development, with effects on both neurons and glia in different brain regions.


Assuntos
Encéfalo/crescimento & desenvolvimento , Histona Desacetilases/metabolismo , Animais , Encéfalo/citologia , Encéfalo/enzimologia , Linhagem Celular Tumoral , Técnicas de Inativação de Genes , Células HEK293 , Histona Desacetilases/deficiência , Histona Desacetilases/genética , Humanos , Camundongos , Neuroglia/metabolismo , Células de Purkinje/metabolismo
11.
J Neurosci ; 33(29): 11833-8, 2013 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-23864673

RESUMO

Huntington's disease (HD) is caused by a polyglutamine expansion within the huntingtin (Htt) protein. Both loss of function of normal Htt and gain of a toxic function by the polyglutamine-expanded mutant Htt protein have been proposed to be responsible for HD, although the molecular mechanisms involved are unclear. We show that Htt is a neuroprotective protein in both HD-related and unrelated model systems. Neuroprotection by Htt is mediated by its sequestration of histone deacetylase-3 (HDAC3), a protein known to promote neuronal death. In contrast to the normal Htt, mutant Htt interacts poorly with HDAC3. However, expression of mutant Htt liberates HDAC3 from Htt, thus de-repressing its neurotoxic activity. Indeed, mutant Htt neurotoxicity is inhibited by the knockdown of HDAC3 and markedly reduced in HDAC3-deficient neurons. A reduction in Htt-HDAC3 interaction is also seen in neurons exposed to other apoptotic stimuli and in the striatum of R6/2 HD mice. Our results suggest that the robust interaction between Htt and HDAC3 along with the ability of mutant Htt to disrupt this association while not itself interacting with HDAC3 provides an explanation for both the loss-of-function and gain-of-toxic-function mechanisms proposed for HD. Moreover, our results identify HDAC3 as an essential player in mutant Htt-induced neurodegeneration.


Assuntos
Corpo Estriado/metabolismo , Proteínas de Ligação a DNA/metabolismo , Histona Desacetilases/metabolismo , Doença de Huntington/metabolismo , Neurônios/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Apoptose/fisiologia , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Histona Desacetilases/genética , Camundongos , Proteínas dos Microfilamentos , Mutação
12.
J Neurosci ; 32(8): 2846-55, 2012 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-22357867

RESUMO

The methyl-CpG binding protein 2 (MeCP2) is a widely expressed protein, the mutations of which cause Rett syndrome. The level of MeCP2 is highest in the brain where it is expressed selectively in mature neurons. Its functions in postmitotic neurons are not known. The MeCP2 gene is alternatively spliced to generate two proteins with different N termini, designated as MeCP2-e1 and MeCP2-e2. The physiological significance of these two isoforms has not been elucidated, and it is generally assumed they are functionally equivalent. We report that in cultured cerebellar granule neurons induced to die by low potassium treatment and in Aß-treated cortical neurons, Mecp2-e2 expression is upregulated whereas expression of the Mecp2-e1 isoform is downregulated. Knockdown of Mecp2-e2 protects neurons from death, whereas knockdown of the e1 isoform has no effect. Forced expression of MeCP2-e2, but not MeCP2-e1, promotes apoptosis in otherwise healthy neurons. We find that MeCP2-e2 interacts with the forkhead protein FoxG1, mutations of which also cause Rett syndrome. FoxG1 has been shown to promote neuronal survival and its downregulation leads to neuronal death. We find that elevated FoxG1 expression inhibits MeCP2-e2 neurotoxicity. MeCP2-e2 neurotoxicity is also inhibited by IGF-1, which prevents the neuronal death-associated downregulation of FoxG1 expression, and by Akt, the activation of which is necessary for FoxG1-mediated neuroprotection. Finally, MeCP2-e2 neurotoxicity is enhanced if FoxG1 expression is suppressed or in neurons cultured from FoxG1-haplodeficient mice. Our results indicate that Mecp2-e2 promotes neuronal death and that this activity is normally inhibited by FoxG1. Reduced FoxG1 expression frees MecP2-e2 to promote neuronal death.


Assuntos
Apoptose/fisiologia , Regulação da Expressão Gênica/genética , Proteína 2 de Ligação a Metil-CpG/metabolismo , Mitose , Neurônios/fisiologia , Síndromes Neurotóxicas/metabolismo , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Apoptose/genética , Células Cultivadas , Cerebelo/citologia , Modelos Animais de Doenças , Feminino , Fatores de Transcrição Forkhead , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína Huntingtina , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Imunoprecipitação , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Proteína 2 de Ligação a Metil-CpG/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Proteínas do Tecido Nervoso/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Síndromes Neurotóxicas/etiologia , Nitrocompostos/toxicidade , Proteínas Nucleares/genética , Potássio/farmacologia , Propionatos/toxicidade , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Transfecção
13.
J Biol Chem ; 287(18): 14749-59, 2012 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-22354967

RESUMO

Transducin-like enhancer of split-1 (TLE1) plays a critical role in the regulation of neurogenesis by inhibiting the differentiation of neural progenitor cells into neurons. Although TLE1 is also expressed highly in the postnatal brain and through adulthood, its role in postmitotic neurons is not clear. Using cultures of cerebellar granule neurons, we show that expression of TLE1 is reduced in neurons primed to die. Reestablishment of elevated TLE1 levels by ectopic expression protects neurons from death, whereas suppression of TLE1 expression in otherwise healthy neurons induces cell death. These results show that TLE1 is necessary for the maintenance of neuronal survival. Experiments using pharmacological inhibitors as well as expression of point mutants indicate that phosphorylation of TLE1 by casein kinase-2 (CK2) at Ser-239 and Ser-253 is necessary for its survival-promoting activity. TLE1-mediated survival is also inhibited by pharmacological inhibition of PI3K-Akt signaling but not by inhibitors of Raf-MEK-ERK signaling or other molecules, including histone deacetylases, calcium calmodulin kinase, or CK1. The survival-promoting activity of TLE1 depends critically on interaction with FoxG1, another protein involved in the regulation of neurogenesis and shown previously to promote survival of postmitotic neurons. Likewise, the ability of FoxG1 to promote neuronal survival depends on TLE1. Taken together, our study demonstrates that TLE1 cooperates with FoxG1 to promote neuronal survival in a CK2- and PI3K-Akt-dependent manner.


Assuntos
Cerebelo/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Proteínas Repressoras/metabolismo , Animais , Morte Celular/fisiologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Cerebelo/citologia , Fatores de Transcrição Forkhead/genética , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas do Tecido Nervoso/genética , Neurônios/citologia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Wistar , Proteínas Repressoras/genética , Quinases raf/genética , Quinases raf/metabolismo
14.
J Biol Chem ; 287(42): 35444-35453, 2012 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-22918830

RESUMO

Both neuroprotective and neurotoxic roles have previously been described for histone deacetylase-1 (HDAC1). Here we report that HDAC1 expression is elevated in vulnerable brain regions of two mouse models of neurodegeneration, the R6/2 model of Huntington disease and the Ca(2+)/calmodulin-dependent protein kinase (CaMK)/p25 double-transgenic model of tauopathic degeneration, suggesting a role in promoting neuronal death. Indeed, elevating HDAC1 expression by ectopic expression promotes the death of otherwise healthy cerebellar granule neurons and cortical neurons in culture. The neurotoxic effect of HDAC1 requires interaction and cooperation with HDAC3, which has previously been shown to selectively induce the death of neurons. HDAC1-HDAC3 interaction is greatly elevated under conditions of neurodegeneration both in vitro and in vivo. Furthermore, the knockdown of HDAC3 suppresses HDAC1-induced neurotoxicity, and the knockdown of HDAC1 suppresses HDAC3 neurotoxicity. As described previously for HDAC3, the neurotoxic effect of HDAC1 is inhibited by treatment with IGF-1, the expression of Akt, or the inhibition of glycogen synthase kinase 3ß (GSK3ß). In addition to HDAC3, HDAC1 has been shown to interact with histone deacetylase-related protein (HDRP), a truncated form of HDAC9, whose expression is down-regulated during neuronal death. In contrast to HDAC3, the interaction between HDRP and HDAC1 protects neurons from death, an effect involving acquisition of the deacetylase activity of HDAC1 by HDRP. We find that elevated HDRP inhibits HDAC1-HDAC3 interaction and prevents the neurotoxic effect of either of these two proteins. Together, our results suggest that HDAC1 is a molecular switch between neuronal survival and death. Its interaction with HDRP promotes neuronal survival, whereas interaction with HDAC3 results in neuronal death.


Assuntos
Ciclo Celular , Córtex Cerebelar/enzimologia , Histona Desacetilase 1/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/enzimologia , Tauopatias/enzimologia , Animais , Proteínas Quinases Dependentes de Cálcio-Calmodulina/genética , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Morte Celular/genética , Córtex Cerebelar/patologia , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Histona Desacetilase 1/genética , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Humanos , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Neurônios/patologia , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Wistar , Tauopatias/genética , Tauopatias/patologia
15.
J Neurosci Res ; 91(3): 407-15, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23239283

RESUMO

Evidence from different laboratories using cell culture and in vivo model systems indicates that histone deacetylase-4 (HDAC4) plays an essential role in maintaining neuronal survival. Indeed, HDAC4 null knockout mice, which die within 2 weeks of birth, display cerebellar degeneration, whereas RNAi-mediated knockdown of HDAC4 expression in the retina of normal mice leads to apoptosis of retinal neurons. As a step toward analyzing the role of HDAC4 in the regulation of neuronal survival in more detail, we generated two separate lines of conditional knockout mice by breeding HDAC4-flox mice with mice expressing Cre recombinase through a Thy1 or nestin promoter. Surprisingly, both Thy1-Cre/HDAC4(-/-) mice, in which HDAC4 is ablated in neurons of the cortex and hippocampus, as well as Nes-Cre/HDAC4(-/-) mice, in which HDAC4 is ablated in neural progenitor cells of the CNS, appear normal at birth, have normal body weight, are fertile, and perform normally in locomotor activity assays. Histological analysis of the brains of Nes-Cre/HDAC4(-/-) mice revealed no obvious abnormalities in cytoarchitecture. Immunohistological analysis of tyrosine hydroxylase and calbindin also showed no discernible defects. Terminal deoxynucleotidyl transferase dUTP nick end-labeling staining showed no difference in the level of neuronal death in the cortex and cerebellum of Nes-Cre/HDAC4(-/-) mice compared with controls. These results indicate that neurons are less dependent on HDAC4 expression for their survival than previously believed and suggest that neuronal death observed in HDAC4 null knockout mice and after RNAi injection may result from HDAC4 deficiency in nonneural cells.


Assuntos
Encéfalo/citologia , Encéfalo/enzimologia , Deleção de Genes , Histona Desacetilases/deficiência , Neurônios/enzimologia , Animais , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Sistema Nervoso Central/citologia , Sistema Nervoso Central/enzimologia , Histona Desacetilases/biossíntese , Histona Desacetilases/genética , Camundongos , Camundongos Knockout
16.
Exp Biol Med (Maywood) ; 248(22): 2095-2108, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-38057990

RESUMO

Rett syndrome is a neurodevelopmental disorder caused by loss-of-function mutations in the methyl-CpG binding protein-2 (MeCP2) gene that is characterized by epilepsy, intellectual disability, autistic features, speech deficits, and sleep and breathing abnormalities. Neurologically, patients with all three disorders display microcephaly, aberrant dendritic morphology, reduced spine density, and an imbalance of excitatory/inhibitory signaling. Loss-of-function mutations in the cyclin-dependent kinase-like 5 (CDKL5) and FOXG1 genes also cause similar behavioral and neurobiological defects and were referred to as congenital or variant Rett syndrome. The relatively recent realization that CDKL5 deficiency disorder (CDD), FOXG1 syndrome, and Rett syndrome are distinct neurodevelopmental disorders with some distinctive features have resulted in separate focus being placed on each disorder with the assumption that distinct molecular mechanisms underlie their pathogenesis. However, given that many of the core symptoms and neurological features are shared, it is likely that the disorders share some critical molecular underpinnings. This review discusses the possibility that deregulation of common molecules in neurons and astrocytes plays a central role in key behavioral and neurological abnormalities in all three disorders. These include KCC2, a chloride transporter, vGlut1, a vesicular glutamate transporter, GluD1, an orphan-glutamate receptor subunit, and PSD-95, a postsynaptic scaffolding protein. We propose that reduced expression or activity of KCC2, vGlut1, PSD-95, and AKT, along with increased expression of GluD1, is involved in the excitatory/inhibitory that represents a key aspect in all three disorders. In addition, astrocyte-derived brain-derived neurotrophic factor (BDNF), insulin-like growth factor 1 (IGF-1), and inflammatory cytokines likely affect the expression and functioning of these molecules resulting in disease-associated abnormalities.


Assuntos
Síndrome de Rett , Espasmos Infantis , Simportadores , Humanos , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Síndrome de Rett/patologia , Mutação , Proteína 4 Homóloga a Disks-Large/genética , Simportadores/genética
17.
J Neurosci ; 31(5): 1746-51, 2011 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-21289184

RESUMO

Although it is well established that pharmacological inhibitors of classical histone deacetylases (HDACs) are protective in various in vivo models of neurodegenerative disease, the identity of the neurotoxic HDAC(s) that these inhibitors target to exert their protective effects has not been resolved. We find that HDAC3 is a protein with strong neurotoxic activity. Forced expression of HDAC3 induces death of otherwise healthy rat cerebellar granule neurons, whereas shRNA-mediated suppression of its expression protects against low-potassium-induced neuronal death. Forced expression of HDAC3 also promotes the death of rat cortical neurons and hippocampally derived HT22 cells, but has no effect on the viability of primary kidney fibroblasts or the HEK293 and HeLa cell lines. This suggests that the toxic effect of HDAC3 is cell selective and that neurons are sensitive to it. Neurotoxicity by HDAC3 is inhibited by treatment with IGF-1 as well as by the expression of a constitutively active form of Akt, an essential mediator of IGF-1 signaling. Protection against HDAC3-induced neurotoxicity is also achieved by the inhibition of GSK3ß, a kinase inhibited by Akt that is widely implicated in the promotion of neurodegeneration in experimental models and in human pathologies. HDAC3 is directly phosphorylated by GSK3ß, suggesting that the neuronal death-promoting action of GSK3ß could be mediated through HDAC3 phosphorylation. In addition to demonstrating that HDAC3 has neurotoxic effects, our study identifies it as a downstream target of GSK3ß.


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , Histona Desacetilases/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Western Blotting , Morte Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebelar/citologia , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Células HEK293 , Células HeLa , Humanos , Imuno-Histoquímica , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/genética , RNA Interferente Pequeno , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transfecção/métodos
18.
J Neurosci ; 31(2): 402-13, 2011 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-21228151

RESUMO

The transcription factor FoxG1 regulates neurogenesis in the embryonic telencephalon as well as a number of other neurodevelopmental processes. While FoxG1 continues to be expressed in neurons postnatally and through adulthood, its role in fully differentiated neurons is not known. The current study demonstrates that FoxG1 promotes the survival of postmitotic neurons. In cerebellar granule neurons primed to undergo apoptosis, FoxG1 expression is reduced. Ectopic expression of FoxG1 blocks neuronal death, whereas suppression of its expression induces death in otherwise healthy neurons. The first 36 residues of FoxG1 are necessary for its survival-promoting effect, while the C-terminal half of the protein is dispensable. Mutation of Asp219, a residue necessary for DNA binding, abrogates survival promotion by FoxG1. Survival promotion is also eliminated by mutation of Thr271, a residue phosphorylated by Akt. Pharmacological inhibition of Akt blocks the survival effects of wild-type FoxG1 but not forms in which Thr271 is replaced with phosphomimetic residues. Treatment of neurons with IGF-1, a neurotrophic factor that promotes neuronal survival by activating Akt, prevents the apoptosis-associated downregulation of FoxG1 expression. Moreover, the overexpression of dominant-negative forms of FoxG1 blocks the ability of IGF-1 to maintain neuronal survival suggesting that FoxG1 is a downstream mediator of IGF-1/Akt signaling. Our study identifies a new and important function for FoxG1 in differentiated neurons.


Assuntos
Fatores de Transcrição Forkhead/fisiologia , Mitose , Proteínas do Tecido Nervoso/fisiologia , Neurônios/citologia , Animais , Apoptose , Diferenciação Celular , Sobrevivência Celular , Células Cultivadas , Cerebelo/citologia , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/genética , Humanos , Fator de Crescimento Insulin-Like I/farmacologia , Fator de Crescimento Insulin-Like I/fisiologia , Camundongos , Mutação , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/fisiologia , Ratos , Ratos Wistar , Transdução de Sinais
19.
J Biol Chem ; 286(6): 4819-28, 2011 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-21118817

RESUMO

Histone deacetylase (HDAC) 7 is a member of the HDAC family of deacetylases. Although some of the HDAC proteins have been shown to regulate neuronal survival and death, whether HDAC7 has a similar role is not known. In this study, we show that HDAC7 protects neurons from apoptosis. In cerebellar granule neurons (CGNs) primed to undergo apoptosis by low potassium treatment, expression of HDAC7 protein is reduced. Reduced expression is also observed in CGNs induced to die by pharmacological inhibition of the proteasome, in cortical neurons treated with homocysteic acid, and in the striatum of R6/2 transgenic mice, a commonly used genetic model of Huntington disease. Forced expression of HDAC7 in cultured CGNs blocks low potassium-induced death, and shRNA-mediated suppression of its expression induces death in otherwise healthy neurons. HDAC7-mediated neuroprotection does not require its catalytic domain and cannot be inhibited by chemical inhibitors of HDACs. Moreover, pharmacological inhibitors of the PI3K-Akt or Raf-MEK-ERK signaling pathways or that of PKA, PKC, and Ca(2+)/calmodulin-dependent protein kinase fail to reduce neuroprotection by HDAC7. We show that stimulation of c-jun expression, an essential feature of neuronal death, is prevented by HDAC7. shRNA-mediated suppression of HDAC7 expression leads to an increase in c-jun expression. Inhibition of c-jun expression by HDAC7 is mediated at the transcriptional level by its direct association with the c-jun gene promoter. Taken together, our results indicate that HDAC7 is a neuroprotective protein acting by a mechanism that is independent of its deacetylase activity but involving the inhibition of c-jun expression.


Assuntos
Cerebelo/metabolismo , Regulação da Expressão Gênica/fisiologia , Histona Desacetilases/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-jun/biossíntese , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Linhagem Celular Tumoral , Cerebelo/citologia , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/genética , MAP Quinase Quinase Quinases/antagonistas & inibidores , MAP Quinase Quinase Quinases/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/citologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/farmacologia , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-jun/genética , Ratos , Ratos Wistar , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/fisiologia
20.
J Neurosci Res ; 88(16): 3508-19, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20936693

RESUMO

Paraneoplastic Ma1 (PNMA1) is a member of a family of proteins involved in an autoimmune disorder called paraneoplastic neurological syndrome. Although it is widely expressed in brain, nothing is known about the function of PNMA1 in neurons. We find that PNMA1 expression is highest in the perinatal brain, a period during which developmentally regulated neuronal death occurs. PNMA1 expression increases in cerebellar granule neurons (CGNs) induced to die by low potassium (LK) and in cortical neurons following homocysteic acid (HCA) treament. Elevated PNMA1 expression is also observed in the degenerating striatum in two separate mouse models of Huntington's disease, the R6/2 transgenic model and the 3-nitropropionic acid-induced chemical model. Suppression of endogenous PNMA1 expression inhibits LK-induced neuronal apoptosis. Ectopic expression of PNMA1 promotes apoptosis even in medium containing high potassium, a condition that normally ensures survival of CGNs. Deletion of the N-terminal half of the PNMA1 protein abrogates its apoptotic activity, whereas deletion of the C-terminal half renders the protein more toxic. Within the N-terminal half, the ability to induce neuronal death depends on the presence of a BH3-like domain. In addition to being necessary for apoptosis, the BH3-like domain is necessary for self-association of PNMA1. Apoptosis by PNMA1 expression is inhibited by overexpression of Bcl2, suggesting that PNMA1-induced neuronal death may depend on the binding of a proapoptotic member of the Bcl2 family to the BH3 domain. Taken together, our results suggest that PNMA1 is a proapoptotic protein in neurons, elevated expression of which may contribute to neurodegenerative disorders.


Assuntos
Antígenos de Neoplasias/metabolismo , Apoptose/fisiologia , Encéfalo/metabolismo , Doença de Huntington/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Animais , Antígenos/metabolismo , Encéfalo/citologia , Encéfalo/crescimento & desenvolvimento , Células Cultivadas , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Mutantes Neurológicos , Camundongos Transgênicos , Degeneração Neural/metabolismo , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA