Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Proc Natl Acad Sci U S A ; 117(8): 4418-4427, 2020 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-32051249

RESUMO

In Alzheimer's disease (AD), human Tau is phosphorylated at S199 (hTau-S199-P) by the protein kinase glycogen synthase kinase 3ß (GSK3ß). HTau-S199-P mislocalizes to dendritic spines, which induces synaptic dysfunction at the early stage of AD. The AKT kinase, once phosphorylated, inhibits GSK3ß by phosphorylating it at S9. In AD patients, the abundance of phosphorylated AKT with active GSK3ß implies that phosphorylated AKT was unable to inactivate GSK3ß. However, the underlying mechanism of the inability of phosphorylated AKT to phosphorylate GSK3ß remains unknown. Here, we show that total AKT and phosphorylated AKT was sulfhydrated at C77 due to the induction of intracellular hydrogen sulfide (H2S). The increase in intracellular H2S levels resulted from the induction of the proinflammatory cytokine, IL-1ß, which is a pathological hallmark of AD. Sulfhydrated AKT does not interact with GSK3ß, and therefore does not phosphorylate GSK3ß. Thus, active GSK3ß phosphorylates Tau aberrantly. In a transgenic knockin mouse (AKT-KI+/+) that lacked sulfhydrated AKT, the interaction between AKT or phospho-AKT with GSK3ß was restored, and GSK3ß became phosphorylated. In AKT-KI+/+ mice, expressing the pathogenic human Tau mutant (hTau-P301L), the hTau S199 phosphorylation was ameliorated as GSK3ß phosphorylation was regained. This event leads to a decrease in dendritic spine loss by reducing dendritic localization of hTau-S199-P, which improves cognitive dysfunctions. Sulfhydration of AKT was detected in the postmortem brains from AD patients; thus, it represents a posttranslational modification of AKT, which primarily contributes to synaptic dysfunction in AD.


Assuntos
Doença de Alzheimer/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Sulfeto de Hidrogênio/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/genética , Motivos de Aminoácidos , Animais , Encéfalo/metabolismo , Feminino , Glicogênio Sintase Quinase 3 beta/genética , Humanos , Interleucina-1beta/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Fosforilação , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt/química , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas tau/genética
2.
J Neuroinflammation ; 19(1): 246, 2022 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-36199097

RESUMO

Differential microglial inflammatory responses play a role in regulation of differentiation and maturation of oligodendrocytes (OLs) in brain white matter. How microglia-OL crosstalk is altered by traumatic brain injury (TBI) and its impact on axonal myelination and neurological function impairment remain poorly understood. In this study, we investigated roles of a Na+/H+ exchanger (NHE1), an essential microglial pH regulatory protein, in microglial proinflammatory activation and OL survival and differentiation in a murine TBI model induced by controlled cortical impact. Similar TBI-induced contusion volumes were detected in the Cx3cr1-CreERT2 control (Ctrl) mice and selective microglial Nhe1 knockout (Cx3cr1-CreERT2;Nhe1flox/flox, Nhe1 cKO) mice. Compared to the Ctrl mice, the Nhe1 cKO mice displayed increased resistance to initial TBI-induced white matter damage and accelerated chronic phase of OL regeneration at 30 days post-TBI. The cKO brains presented increased anti-inflammatory phenotypes of microglia and infiltrated myeloid cells, with reduced proinflammatory transcriptome profiles. Moreover, the cKO mice exhibited accelerated post-TBI sensorimotor and cognitive functional recovery than the Ctrl mice. These phenotypic outcomes in cKO mice were recapitulated in C57BL6J wild-type TBI mice receiving treatment of a potent NHE1 inhibitor HOE642 for 1-7 days post-TBI. Taken together, these findings collectively demonstrated that blocking NHE1 protein stimulates restorative microglial activation in oligodendrogenesis and neuroprotection, which contributes to accelerated brain repair and neurological function recovery after TBI.


Assuntos
Lesões Encefálicas Traumáticas , Substância Branca , Animais , Lesões Encefálicas Traumáticas/metabolismo , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Oligodendroglia , Recuperação de Função Fisiológica
3.
Mol Cell ; 56(6): 786-95, 2014 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-25435139

RESUMO

Induction of a proinflammatory cytokine, interleukin-1ß (IL-1ß) plays a role in memory impairment associated with various neurological disorders and brain injury. Here we show that IL-1ß-induced memory impairment in brain is mediated by hydrogen sulfide (H2S) synthesized by cystathionine beta-synthase (CBS). H2S modifies GAPDH essentially via sulfhydration in dendrites, which promotes its binding to the E3 ligase protein, Siah. Then Siah binds to a critical synaptic scaffolding molecule, PSD95, and leads it to degradation via ubiquitination. In CBS heterozygous mice (cbs(+/-)) and primary neurons depleted with either CBS or IL-1R, IL-1ß-induced loss of PSD95 was rescued along with a decrease in the level of GAPDH sulfhydration. Moreover, decrease in the loss of PSD95 in cbs(+/-) mice results in improvement of IL-1ß-induced cognitive deficits and neurobehavioral outcomes. Thus, our findings reveal a mechanism where GAPDH sulfhydration appears to be a physiologic determinant of cytokine-induced memory impairment in brain.


Assuntos
Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Guanilato Quinases/metabolismo , Interleucina-1beta/fisiologia , Proteínas de Membrana/metabolismo , Animais , Cistationina beta-Sintase/metabolismo , Proteína 4 Homóloga a Disks-Large , Células HEK293 , Humanos , Sulfeto de Hidrogênio/metabolismo , Masculino , Transtornos da Memória/enzimologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Processamento de Proteína Pós-Traducional , Proteólise , Sinapses/enzimologia , Sinapses/patologia
4.
J Neurosci ; 40(2): 424-446, 2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31694961

RESUMO

Persistent endoplasmic reticulum (ER) stress in neurons is associated with activation of inflammatory cells and subsequent neuroinflammation following traumatic brain injury (TBI); however, the underlying mechanism remains elusive. We found that induction of neuronal-ER stress, which was mostly characterized by an increase in phosphorylation of a protein kinase R-like ER kinase (PERK) leads to release of excess interferon (IFN)ß due to atypical activation of the neuronal-STING signaling pathway. IFNß enforced activation and polarization of the primary microglial cells to inflammatory M1 phenotype with the secretion of a proinflammatory chemokine CXCL10 due to activation of STAT1 signaling. The secreted CXCL10, in turn, stimulated the T-cell infiltration by serving as the ligand and chemoattractant for CXCR3+ T-helper 1 (Th1) cells. The activation of microglial cells and infiltration of Th1 cells resulted in white matter injury, characterized by impaired myelin basic protein and neurofilament NF200, the reduced thickness of corpus callosum and external capsule, and decline of mature oligodendrocytes and oligodendrocyte precursor cells. Intranasal delivery of CXCL10 siRNA blocked Th1 infiltration but did not fully rescue microglial activation and white matter injury after TBI. However, impeding PERK-phosphorylation through the administration of GSK2656157 abrogated neuronal induction of IFNß, switched microglial polarization to M2 phenotype, prevented Th1 infiltration, and increased Th2 and Treg levels. These events ultimately attenuated the white matter injury and improved anxiety and depressive-like behavior following TBI.SIGNIFICANCE STATEMENT A recent clinical study showed that human brain trauma patients had enhanced expression of type-1 IFN; suggests that type-1 IFN signaling may potentially influence clinical outcome in TBI patients. However, it was not understood how TBI leads to an increase in IFNß and whether induction of IFNß has any influence on neuroinflammation, which is the primary reason for morbidity and mortality in TBI. Our study suggests that induction of PERK phosphorylation, a characteristic feature of ER stress is responsible for an increase in neuronal IFNß, which, in turn, activates microglial cells and subsequently manifests the infiltration of T cells to induce neuroinflammation and subsequently white matter injury. Blocking PERK phosphorylation using GSK2656157 (or PERK knockdown) the whole cascade of neuroinflammation was attenuated and improved cognitive function after TBI.


Assuntos
Lesões Encefálicas Traumáticas/fisiopatologia , Estresse do Retículo Endoplasmático/fisiologia , Microglia/metabolismo , Linfócitos T , Substância Branca/fisiopatologia , eIF-2 Quinase/metabolismo , Animais , Feminino , Interferon beta/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Transdução de Sinais/fisiologia , Substância Branca/lesões
5.
Mol Cell ; 45(1): 13-24, 2012 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-22244329

RESUMO

Nuclear factor κB (NF-κB) is an antiapoptotic transcription factor. We show that the antiapoptotic actions of NF-κB are mediated by hydrogen sulfide (H(2)S) synthesized by cystathionine gamma-lyase (CSE). TNF-α treatment triples H(2)S generation by stimulating binding of SP1 to the CSE promoter. H(2)S generated by CSE stimulates DNA binding and gene activation of NF-κB, processes that are abolished in CSE-deleted mice. As CSE deletion leads to decreased glutathione levels, resultant oxidative stress may contribute to alterations in CSE mutant mice. H(2)S acts by sulfhydrating the p65 subunit of NF-κB at cysteine-38, which promotes its binding to the coactivator ribosomal protein S3 (RPS3). Sulfhydration of p65 predominates early after TNF-α treatment, then declines and is succeeded by a reciprocal enhancement of p65 nitrosylation. In CSE mutant mice, antiapoptotic influences of NF-κB are markedly diminished. Thus, sulfhydration of NF-κB appears to be a physiologic determinant of its antiapoptotic transcriptional activity.


Assuntos
Apoptose/fisiologia , Sulfeto de Hidrogênio/química , NF-kappa B/química , Animais , Cistationina gama-Liase/genética , Cistationina gama-Liase/metabolismo , Cistationina gama-Liase/fisiologia , Regulação da Expressão Gênica , Camundongos , NF-kappa B/fisiologia , Fator de Transcrição Sp1/metabolismo , Fator de Transcrição RelA/química , Fator de Transcrição RelA/metabolismo , Fator de Necrose Tumoral alfa/fisiologia
6.
J Neurosci ; 37(24): 5900-5911, 2017 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-28522733

RESUMO

The PKR-like ER kinase (PERK), a transmembrane protein, resides in the endoplasmic reticulum (ER). Its activation serves as a key sensor of ER stress, which has been implicated in traumatic brain injury (TBI). The loss of memory is one of the most common symptoms after TBI, but the precise role of PERK activation in memory impairment after TBI has not been well elucidated. Here, we have shown that blocking the activation of PERK using GSK2656157 prevents the loss of dendritic spines and rescues memory deficits after TBI. To elucidate the molecular mechanism, we found that activated PERK phosphorylates CAMP response element binding protein (CREB) and PSD95 directly at the S129 and T19 residues, respectively. Phosphorylation of CREB protein prevents its interaction with a coactivator, CREB-binding protein, and subsequently reduces the BDNF level after TBI. Conversely, phosphorylation of PSD95 leads to its downregulation in pericontusional cortex after TBI in male mice. Treatment with either GSK2656157 or overexpression of a kinase-dead mutant of PERK (PERK-K618A) rescues BDNF and PSD95 levels in the pericontusional cortex by reducing phosphorylation of CREB and PSD95 proteins after TBI. Similarly, administration of either GSK2656157 or overexpression of PERK-K618A in primary neurons rescues the loss of dendritic outgrowth and number of synapses after treatment with a PERK activator, tunicamycin. Therefore, our study suggests that inhibition of PERK phosphorylation could be a potential therapeutic target to restore memory deficits after TBI.SIGNIFICANCE STATEMENT Traumatic brain injury (TBI) is the leading cause of death and disability around the world and affects 1.7 million Americans each year. Here, we have shown that TBI-activated PKR-like ER kinase (PERK) is responsible for memory deficiency, which is the most common problem in TBI patients. A majority of PERK's biological activities have been attributed to its function as an eIF2α kinase. However, our study suggests that activated PERK mediates its function via increasing phosphorylation of CAMP response element binding protein (CREB) and PSD95 after TBI. Blocking PERK phosphorylation rescues spine loss and memory deficits independently of phosphorylation of eIF2α. Therefore, our study suggests that CREB and PSD95 are novel substrates of PERK, so inhibition of PERK phosphorylation using GSK2656157 would be beneficial against memory impairment after TBI.


Assuntos
Lesões Encefálicas Traumáticas/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Guanilato Quinases/metabolismo , Proteínas de Membrana/metabolismo , Transtornos da Memória/metabolismo , eIF-2 Quinase/metabolismo , Animais , Lesões Encefálicas Traumáticas/complicações , Lesões Encefálicas Traumáticas/patologia , Espinhas Dendríticas/patologia , Proteína 4 Homóloga a Disks-Large , Regulação para Baixo , Ativação Enzimática , Masculino , Transtornos da Memória/etiologia , Transtornos da Memória/patologia , Camundongos , Camundongos Endogâmicos C57BL
7.
Neurobiol Dis ; 118: 108-116, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30010002

RESUMO

Cell cycle activation has been associated with varying types of neurological disorders including brain injury. Cyclin D1 is a critical modulator of cell cycle activation and upregulation of Cyclin D1 in neurons contributes to the pathology associated with traumatic brain injury (TBI). Mitochondrial mass is a critical factor to maintain the mitochondrial function, and it can be regulated by different signaling cascades and transcription factors including NRF1. However, the underlying mechanism of how TBI leads to impairment of mitochondrial mass following TBI remains obscure. Our results indicate that augmentation of CyclinD1 attenuates mitochondrial mass formation following TBI. To elucidate the molecular mechanism, we found that Cyclin D1 interacts with a transcription factor NRF1 in the nucleus and prevents NRF1's interaction with p300 in the pericontusional cortex following TBI. As a result, the acetylation level of NRF1 was decreased, and its transcriptional activity was attenuated. This event leads to a loss of mitochondrial mass in the pericontusional cortex following TBI. Intranasal delivery of Cyclin D1 RNAi immediately after TBI rescues transcriptional activation of NRF1 and recovers mitochondrial mass after TBI.


Assuntos
Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/patologia , Ciclina D1/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Animais , Lesões Encefálicas Traumáticas/genética , Ciclina D1/antagonistas & inibidores , Ciclina D1/genética , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/genética , Interferência de RNA/fisiologia
8.
Neurobiol Dis ; 96: 12-21, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27544482

RESUMO

Anesthetics including isoflurane are known to induce neuronal dysfunction in the developing brain, however, the underlying mechanism is mostly unknown. The transcriptional activation of CREB (cyclic AMP response element binding protein) and the alterations in acetylation of histones modulated by several histone deacetylases such as HDAC4 (histone deacetylase 4) are known to contribute to synaptic plasticity in the brain. Here we have shown that administration of isoflurane (1.4%) for 2h leads to transcriptional inactivation of CREB which results in loss of dendritic outgrowth and decreased expression level of proteins essential for memory and cognitive functions, such as BDNF, and c-fos in the developing brain of mice at postnatal day 7 (PND7). To elucidate the molecular mechanism, we found that exposure to isoflurane leads to an increase in nuclear translocation of HDAC4, which interacts with CREB in the nucleus. This event, in turn, results in a decrease in interaction between an acetyltransferase, CBP, and CREB that ultimately leads to transcriptional inactivation of CREB. As a result, the expression level of BDNF, and c-fos were significantly down-regulated after administration of isoflurane in PND7 brain. Depletion of HDAC4 in PND7 brain rescues the transcriptional activation of CREB along with augmentation in the level of the expression level of BDNF and c-fos. Moreover, administration of lentiviral particles of HDAC4 RNAi in primary neurons rescues neurite outgrowth following isoflurane treatment. Taken together, our study suggests that HDAC4-induced transcriptional inactivation of CREB is responsible for isoflurane-induced cognitive dysfunction in the brain.


Assuntos
Anestésicos Inalatórios/toxicidade , Encéfalo , Proteína de Ligação a CREB/metabolismo , Transtornos Cognitivos/induzido quimicamente , Histona Desacetilases/metabolismo , Isoflurano/toxicidade , Animais , Animais Recém-Nascidos , Encéfalo/efeitos dos fármacos , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Encéfalo/patologia , Transtornos Cognitivos/patologia , Transtornos Cognitivos/fisiopatologia , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , Histona Desacetilases/genética , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Crescimento Neuronal/efeitos dos fármacos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Reflexo de Endireitamento/efeitos dos fármacos
9.
Proc Natl Acad Sci U S A ; 110(40): 16181-6, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-24043835

RESUMO

Profound induction of immediate early genes (IEGs) by neural activation is a critical determinant for plasticity in the brain, but intervening molecular signals are not well characterized. We demonstrate that inositol polyphosphate multikinase (IPMK) acts noncatalytically as a transcriptional coactivator to mediate induction of numerous IEGs. IEG induction by electroconvulsive stimulation is virtually abolished in the brains of IPMK-deleted mice, which also display deficits in spatial memory. Neural activity stimulates binding of IPMK to the histone acetyltransferase CBP and enhances its recruitment to IEG promoters. Interestingly, IPMK regulation of CBP recruitment and IEG induction does not require its catalytic activities. Dominant-negative constructs, which prevent IPMK-CBP binding, substantially decrease IEG induction. As IPMK is ubiquitously expressed, its epigenetic regulation of IEGs may influence diverse nonneural and neural biologic processes.


Assuntos
Encéfalo/metabolismo , Epigênese Genética/fisiologia , Regulação da Expressão Gênica/fisiologia , Genes Precoces/fisiologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Ativação Transcricional/fisiologia , Análise de Variância , Animais , Proteína de Ligação a CREB/metabolismo , Imunoprecipitação da Cromatina , Regulação da Expressão Gênica/genética , Genes Precoces/genética , Aprendizagem em Labirinto , Camundongos , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Reconhecimento Psicológico/fisiologia
10.
J Neurosci ; 33(44): 17398-403, 2013 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-24174672

RESUMO

Increased cranial pressure due to development of edema contributes significantly to the pathology of traumatic brain injury (TBI). Induction of an astrocytic water channel protein, Aquaporin 4 (AQP4), is known to predominantly contribute to cytotoxic edema following TBI. However, the mechanism for the increase in AQP4 following 24 h of TBI is poorly understood. Here we show that transcriptional activation of a ubiquitously expressed mammalian forkhead transcription factor, Foxo3a, induces cerebral edema by increasing the AQP4 level in the controlled cortical impact model of TBI in mice. TBI stimulates nuclear translocation of Foxo3a in astrocytes and subsequently augments its binding to AQP4 promoter in pericontusional cortex. Nuclear accumulation of Foxo3a is augmented by a decrease in phosphorylation at its Ser256 residue due to inactivation of Akt after TBI. Depletion of Foxo3a in mice rescues cytotoxic edema by preventing induction of AQP4 as well as attenuates memory impairment after TBI in mice.


Assuntos
Aquaporina 4/biossíntese , Edema Encefálico/etiologia , Lesões Encefálicas/metabolismo , Fatores de Transcrição Forkhead/fisiologia , Regulação para Cima/fisiologia , Transporte Ativo do Núcleo Celular/genética , Animais , Aquaporina 4/genética , Sequência de Bases , Edema Encefálico/genética , Edema Encefálico/patologia , Lesões Encefálicas/genética , Lesões Encefálicas/patologia , Células Cultivadas , Modelos Animais de Doenças , Regulação para Baixo/genética , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Fosforilação/genética , Transcrição Gênica , Regulação para Cima/genética
11.
Proc Natl Acad Sci U S A ; 108(50): 20178-83, 2011 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-22123949

RESUMO

Epigenetic regulation of histones mediates neurotrophin actions with histone acetylation enhancing cAMP response element-binding (CREB)-associated transcription elicited by brain-derived neurotrophic factor (BDNF) and nerve-growth factor (NGF). Roles for histone methylation in CREB's transcriptional activity have not been well characterized. We show that depletion of the histone methyltransferase suppressor of variegation 3-9 homolog 1 (SUV39H1) selectively augments BDNF- and NGF-mediated neurite outgrowth. SUV39H1 is the principal enzyme responsible for trimethylation of histone H3 at lysine 9, a molecular mark associated with transcriptional silencing. BDNF and NGF act via a signaling cascade wherein degradation of SUV39H1 down-regulates trimethylation of H3K9 in a nitric oxide-dependent pathway. BDNF activates neuronal NOS with the nitrosylated GAPDH/seven in absentia (Siah) homolog complex translocating to the nucleus. Degradation of SUV39H1 by Siah facilitates histone H3 on lysine 9 acetylation, CREB binding to DNA, enhanced expression of CREB-regulated genes and neurite outgrowth.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Histona-Lisina N-Metiltransferase/metabolismo , Fatores de Crescimento Neural/farmacologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Dendritos/metabolismo , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/metabolismo , Histona Metiltransferases , Histonas/metabolismo , Metilação/efeitos dos fármacos , Camundongos , Neurônios/enzimologia , Óxido Nítrico/metabolismo , Nitrosação/efeitos dos fármacos , Células PC12 , Ligação Proteica/efeitos dos fármacos , Estabilidade Proteica/efeitos dos fármacos , Ratos , Ubiquitinação/efeitos dos fármacos
12.
J Biol Chem ; 286(50): 43204-13, 2011 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-22020940

RESUMO

Tumor protein (TP)-p53 family members often play proapoptotic roles, whereas nuclear factor κB (NF-κB) functions as a proapoptotic and antiapoptotic regulator depending on the cellular environment. We previously showed that the NF-κB activation leads to the reduction of the TP63 isoform, ΔNp63α, thereby rendering the cells susceptible to cell death upon DNA damage. However, the functional relationship between TP63 isotypes and NF-κB is poorly understood. Here, we report that the TAp63 regulates NF-κB transcription and protein stability subsequently leading to the cell death phenotype. We found that TAp63α induced the expression of the p65 subunit of NF-κB (RELA) and target genes involved in cell cycle arrest or apoptosis, thereby triggering cell death pathways in MCF10A cells. RELA was shown to concomitantly modulate specific cell survival pathways, making it indispensable for the TAp63α-dependent regulation of cell death. We showed that TAp63α and RELA formed protein complexes resulted in their mutual stabilization and inhibition of the RELA ubiquitination. Finally, we showed that TAp63α directly induced RelA transcription by binding to and activating of its promoter and, in turn, leading to activation of the NF-κB-dependent cell death genes. Overall, our data defined the regulatory feedback loop between TAp63α and NF-κB involved in the activation of cell death process of cancer cells.


Assuntos
Fator de Transcrição RelA/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Morte Celular/genética , Morte Celular/fisiologia , Linhagem Celular , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Imunoprecipitação da Cromatina , Humanos , Immunoblotting , Imunoprecipitação , Ligação Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição RelA/genética , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética
13.
Mol Microbiol ; 78(1): 119-37, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20659295

RESUMO

Tyrosyl DNA phosphodiesterase 1 (Tdp1) is a member of phospholipase D superfamily, which cleaves a broad range of 3'-DNA adducts, the best characterized of which is the phosphodiester bond formed between DNA and topoisomerase IB. This study describes cloning and functional characterization of the enzyme, termed as LdTdp1 in the kinetoplastid parasite Leishmania donovani. Sequence analysis confirmed conservation of the active site motifs typical for all Tdp1 proteins. LdTdp1 activity was detected in the parasite nucleus as well as in the kinetoplast. The enzyme harbours a nuclear localization signal at its C-terminus. Overexpression of the active enzyme protected the parasites against topoisomerase IB inhibitor camptothecin (CPT) and oxidative agent H(2)O(2)-mediated cytotoxicity and its downregulation rendered the parasites hypersensitive to CPT. Trapping of mutant LdTdp1 on DNA takes place following CPT treatment in L. donovani cells. The expression level and associated activity of LdTdp1 were found to be higher in CPT-resistant L. donovani parasites. Altogether, this is the first report of Tdp1 from the kinetoplastid parasite L. donovani, which actively participates in topoisomerase I-mediated DNA damage repair process and thereby counteracts the cytotoxic effect of topoisomerase I inhibitors.


Assuntos
Reparo do DNA , DNA de Protozoário/metabolismo , Leishmania donovani/enzimologia , Diester Fosfórico Hidrolases/metabolismo , Proteínas de Protozoários/metabolismo , Apoptose , Camptotecina/farmacologia , Núcleo Celular/genética , Núcleo Celular/metabolismo , Clonagem Molecular , Técnicas de Silenciamento de Genes , Peróxido de Hidrogênio/farmacologia , Leishmania donovani/genética , Modelos Moleculares , Mutagênese Sítio-Dirigida , Estresse Oxidativo , Diester Fosfórico Hidrolases/genética , Plastídeos/genética , Plastídeos/metabolismo , Proteínas de Protozoários/genética , Alinhamento de Sequência
14.
Neuroscientist ; 25(5): 420-433, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30477403

RESUMO

The brain undergoes several changes at structural, molecular, and cellular levels leading to alteration in its functions and these processes are primarily maintained by proteostasis in cells. However, an imbalance in proteostasis due to the abnormal accumulation of protein aggregates induces endoplasmic reticulum (ER) stress. This event, in turn, activate the unfolded protein response; however, in most neurodegenerative conditions and brain injury, an uncontrolled unfolded protein response elicits memory dysfunction. Although the underlying signaling mechanism for impairment of memory function following induction of ER stress remains elusive, recent studies have highlighted that inactivation of a transcription factor, CREB, which is essential for synaptic function and memory formation, plays an essential role for ER stress-induced synaptic and memory dysfunction. In this review, current studies and most updated view on how ER stress affects memory function in both physiological and pathological conditions will be highlighted.


Assuntos
Lesões Encefálicas/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Memória/fisiologia , Proteostase/fisiologia , Resposta a Proteínas não Dobradas/fisiologia , Animais , Lesões Encefálicas/complicações , Humanos , Transtornos da Memória/etiologia , Transtornos da Memória/metabolismo , Neurônios/metabolismo , Transdução de Sinais
15.
Mech Ageing Dev ; 178: 72-79, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30668956

RESUMO

Tau, a microtubule-associated protein promotes assembly and stability of microtubules which is related to axoplasmic flow and critical neuronal activities upon physiological conditions. Under neurodegenerative condition such as in Alzheimer's Disease (AD), tau-microtubule binding dynamics and equilibrium are severely affected due to its aberrant post-translational modifications including acetylation and hyperphosphorylation. This event results in its conformational changes to form neurofibrillary tangles (NFT) after aggregation in the cytosol. The formation of NFT is more strongly correlated with cognitive decline than the distribution of senile plaque, which is formed by polymorphous beta-amyloid (Aß) protein deposits, another pathological hallmark of AD. In neurodegenerative conditions, other than AD, the disease manifestation is correlated with mutations of the MAPT gene. In Primary age-related tauopathy (PART), which is commonly observed in the brains of aged individuals, tau deposition is directly correlated with cognitive deficits even in the absence of Aß deposition. Thus, tauopathy has been considered as an essential hallmark in neurodegeneration and normal brain aging. In this review, we highlighted the recent progress about the tauopathies in the light of its posttranslational modifications and its implication in AD and the aged brain.


Assuntos
Envelhecimento/metabolismo , Encéfalo/metabolismo , Processamento de Proteína Pós-Traducional , Tauopatias/metabolismo , Proteínas tau/metabolismo , Acetilação , Envelhecimento/patologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Encéfalo/patologia , Disfunção Cognitiva/metabolismo , Humanos , Emaranhados Neurofibrilares/metabolismo , Estresse Oxidativo/fisiologia , Fosforilação , Placa Amiloide/fisiopatologia , Tauopatias/patologia
16.
Free Radic Biol Med ; 134: 630-643, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30790655

RESUMO

Traumatic Brain Injury (TBI) affects more than 1.7 million Americans each year and about 30% of TBI-patients having visual impairments. The loss of retinal ganglion cells (RGC) in the retina and axonal degeneration in the optic nerve have been attributed to vision impairment following TBI; however, the molecular mechanism has not been elucidated. Here we have shown that an increase in histone di-methylation at lysine 9 residue (H3K9Me2), synthesized by the catalytic activity of a histone methyltransferase, G9a is responsible for RGC loss and axonal degeneration in the optic nerve following TBI. To elucidate the molecular mechanism, we found that an increase in H3K9Me2 results in the induction of oxidative stress both in the RGC and optic nerve by decreasing the mRNA level of antioxidants such as Superoxide dismutase (sod) and catalase through impairing the transcriptional activity of Nuclear factor E2-related factor 2 (Nrf2) via direct interaction. The induction of oxidative stress is associated with death in RGC and oligodendrocyte precursor cells (OPCs). The death in OPCs is correlated with a reduction in myelination, and the expression of myelin binding protein (MBP) in association with degeneration of neurofilaments in the optic nerve. This event allied to an impairment of the retrograde transport of axons and loss of nerve fiber layer in the optic nerve following TBI. An administration of G9a inhibitor, UNC0638 attenuates the induction of H3K9Me2 both in RGC and optic nerve and subsequently activates Nrf2 to reduce oxidative stress. This event was concomitant with the rescue in the loss of retinal thickness, attenuation in optic nerve degeneration and improvement in the retrograde transport of axons following TBI.


Assuntos
Lesões Encefálicas Traumáticas/complicações , Histonas/metabolismo , Lisina/metabolismo , Nervo Óptico/patologia , Estresse Oxidativo , Células Ganglionares da Retina/patologia , Transtornos da Visão/etiologia , Animais , Lesões Encefálicas Traumáticas/patologia , Modelos Animais de Doenças , Histonas/genética , Lisina/genética , Masculino , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Bainha de Mielina , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Nervo Óptico/metabolismo , Células Ganglionares da Retina/metabolismo , Transtornos da Visão/metabolismo , Transtornos da Visão/patologia
17.
J Neurotrauma ; 36(23): 3284-3296, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31169064

RESUMO

An enduring deficit in neurogenesis largely contributes to the development of severe post-traumatic psychiatric disorders such as anxiety, depression, and memory impairment following traumatic brain injury (TBI); however, the mechanism remains obscure. Here we have shown that an imbalance in the generation of γ-aminobutyric acid (GABA)ergic and glutamatergic neurons due to aberrant induction of vesicular glutamate transporter 1 (vGlut1)-positive glutamatergic cells is responsible for impaired neuronal differentiation in the hippocampus following TBI. To elucidate the molecular mechanism, we found that TBI activates a transcription factor, Pax3, by increasing its acetylation status, and subsequently induces Ngn2 transcription. This event, in turn, augments the vGlut1-expressing glutamatergic neurons and accumulation of excess glutamate in the hippocampus that can affect neuronal differentiation. In our study the acetylation of Pax3 was increased due to loss of its interaction with a deacetylase, histone deacetylase 4 (HDAC4), which was downregulated after TBI. TBI-induced activation of GSK3ß was responsible for the degradation of HDAC4. We also showed that overexpression of HDAC4 before TBI reduces Pax3 acetylation by restoring an interaction between HDAC4 and Pax3 in the hippocampus. This event prevents the aberrant induction of vGlut1-positive glutamatergic neurons by decreasing the Ngn2 level and subsequently reinforces the balance between GABAergic and glutamatergic neurons following TBI. Further, we found that overexpression of HDAC4 in the hippocampus improves anxiety, depressive-like behavior, and memory functions following TBI.


Assuntos
Lesões Encefálicas Traumáticas/enzimologia , Regulação para Baixo/fisiologia , Histona Desacetilases/metabolismo , Transtornos Mentais/enzimologia , Neurogênese/fisiologia , Animais , Lesões Encefálicas Traumáticas/patologia , Lesões Encefálicas Traumáticas/psicologia , Feminino , Masculino , Aprendizagem em Labirinto/fisiologia , Transtornos Mentais/patologia , Transtornos Mentais/psicologia , Camundongos , Camundongos Endogâmicos C57BL
18.
Nucleic Acids Res ; 34(21): 6286-97, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17098934

RESUMO

The active site tyrosine residue of all monomeric type IB topoisomerases resides in the C-terminal domain of the enzyme. Leishmania donovani, possesses unusual heterodimeric type IB topoisomerase. The small subunit harbors the catalytic tyrosine within the SKXXY motif. To explore the functional relationship between the two subunits, we have replaced the small subunit of L.donovani topoisomerase I with a C-terminal fragment of human topoisomerase I (HTOP14). The purified LdTOP1L (large subunit of L.donovani topoisomerase I) and HTOP14 were able to reconstitute topoisomerase I activity when mixed in vitro. This unusual enzyme, 'LeishMan' topoisomerase I (Leish for Leishmania and Man for human) exhibits less efficiency in DNA binding and strand passage compared with LdTOP1L/S. Fusion of LdTOP1L with HTOP14 yielded a more efficient enzyme with greater affinity for DNA and faster strand passage ability. Both the chimeric enzymes are less sensitive to camptothecin than LdTOP1L/S. Restoration of topoisomerase I activity by LdTOP1L and HTOP14 suggests that the small subunit of L.donovani topoisomerase I is primarily required for supplying the catalytic tyrosine. Moreover, changes in the enzyme properties due to substitution of LdTOP1S with HTOP14 indicate that the small subunit contributes to subunit interaction and catalytic efficiency of the enzyme.


Assuntos
DNA Topoisomerases Tipo I/metabolismo , Leishmania donovani/enzimologia , Proteínas de Protozoários/metabolismo , Animais , Camptotecina/farmacologia , DNA/metabolismo , DNA Topoisomerases Tipo I/química , DNA Topoisomerases Tipo I/genética , Polarização de Fluorescência , Potássio/farmacologia , Subunidades Proteicas/genética , Subunidades Proteicas/fisiologia , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Proteínas Recombinantes de Fusão/metabolismo
19.
Nucleic Acids Res ; 34(4): 1121-32, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16488884

RESUMO

Emergence of the bi-subunit topoisomerase I in the kinetoplastid family (Trypanosoma and Leishmania) has brought a new twist in topoisomerase research related to evolution, functional conservation and preferential sensitivities to the specific inhibitors of type IB topoisomerase family. In the present study, we describe that naturally occurring flavones baicalein, luteolin and quercetin are potent inhibitors of the recombinant Leishmania donovani topoisomerase I. These compounds bind to the free enzyme and also intercalate into the DNA at a very high concentration (300 microM) without binding to the minor grove. Here, we show that inhibition of topoisomerase I by these flavones is due to stabilization of topoisomerase I-DNA cleavage complexes, which subsequently inhibit the religation step. Their ability to stabilize the covalent topoisomerase I-DNA complex in vitro and in living cells is similar to that of the known topoisomerase I inhibitor camptothecin (CPT). However, in contrast to CPT, baicalein and luteolin failed to inhibit the religation step when the drugs were added to pre-formed enzyme substrate binary complex. This differential mechanism to induce the stabilization of cleavable complex with topoisomerase I and DNA by these selected flavones and CPT led us to investigate the effect of baicalein and luteolin on CPT-resistant mutant enzyme LdTOP1Delta39LS lacking 1-39 amino acids of the large subunit [B. B. Das, N. Sen, S. B. Dasgupta, A. Ganguly and H. K. Majumder (2005) J. Biol. Chem. 280, 16335-16344]. Baicalein and luteolin stabilize duplex oligonucleotide cleavage with LdTOP1Delta39LS. This observation was further supported by the stabilization of in vivo cleavable complex by baicalein and luteolin with highly CPT-resistant L.donovani strain. Taken together, our data suggest that the interacting amino acid residues of topoisomerase I may be partially overlapping or different for flavones and CPT. This study illuminates new properties of the flavones and provide additional insights into the ligand binding properties of L.donovani topoisomerase I.


Assuntos
Camptotecina/farmacologia , DNA Topoisomerases Tipo I/efeitos dos fármacos , DNA/metabolismo , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Leishmania donovani/enzimologia , Animais , DNA/química , DNA Topoisomerases Tipo I/genética , DNA Topoisomerases Tipo I/metabolismo , Flavanonas/química , Flavanonas/farmacologia , Flavonoides/química , Luteolina/química , Luteolina/farmacologia , Mutação , Subunidades Proteicas/metabolismo , Proteínas de Protozoários/efeitos dos fármacos , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Quercetina/química , Quercetina/farmacologia
20.
Sci Signal ; 11(522)2018 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-29559585

RESUMO

Acetylation of the microtubule-associated protein tau promotes its polymerization into neurofibrillary tangles that are implicated in the pathology of Alzheimer's disease (AD). The gaseous neurotransmitter nitric oxide (NO) regulates cell signaling through the nitrosylation of proteins. We found that NO production and tau acetylation at Lys280 occurred in the brain tissue in mice and in cultured mouse cortical neurons in response to exposure to amyloid-ß1-42 (Aß1-42), a peptide that is also implicated in AD. An increased abundance of NO facilitated the S-nitrosylation (SNO) of glyceraldehyde-3-phosphate dehydrogenase (GAPDH). S-nitrosylated GAPDH (GAPDH-SNO) promoted the acetylation and activation of the acetyltransferase p300 and facilitated the nitrosylation and inactivation of the deacetylase sirtuin 1 (SIRT1). The abundance of GAPDH-SNO was increased in postmortem brain samples from AD patients. Preventing the increase in GAPDH-SNO abundance in both cultured neurons and mice, either by overexpression of the nitrosylation mutant of GAPDH (GAPDH C150S) or by treatment with the GAPDH nitrosylation inhibitor CGP3466B (also known as omigapil), abrogated Aß1-42-induced tau acetylation, memory impairment, and locomotor dysfunction in mice, suggesting that this drug might be repurposed to treat patients with AD.


Assuntos
Doença de Alzheimer/metabolismo , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Óxido Nítrico Sintase Tipo I/deficiência , Óxido Nítrico/metabolismo , Sirtuína 1/metabolismo , Fatores de Transcrição de p300-CBP/metabolismo , Proteínas tau/metabolismo , Acetilação/efeitos dos fármacos , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/toxicidade , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Óxido Nítrico Sintase Tipo I/genética , Oxepinas/farmacologia , Fragmentos de Peptídeos/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA