Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(11)2023 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-37298071

RESUMO

The apolipoprotein E (ApoE) gene is a genetic risk factor for late-onset Alzheimer's disease, in which ε4 allele carriers have increased risk compared to the common ε3 carriers. Cadmium (Cd) is a toxic heavy metal and a potential neurotoxicant. We previously reported a gene-environment interaction (GxE) effect between ApoE4 and Cd that accelerates or increases the severity of the cognitive decline in ApoE4-knockin (ApoE4-KI) mice exposed to 0.6 mg/L CdCl2 through drinking water compared to control ApoE3-KI mice. However, the mechanisms underlying this GxE effect are not yet defined. Because Cd impairs adult neurogenesis, we investigated whether genetic and conditional stimulation of adult neurogenesis can functionally rescue Cd-induced cognitive impairment in ApoE4-KI mice. We crossed either ApoE4-KI or ApoE3-KI to an inducible Cre mouse strain, Nestin-CreERTM:caMEK5-eGFPloxP/loxP (designated as caMEK5), to generate ApoE4-KI:caMEK5 and ApoE3-KI:caMEK5. Tamoxifen administration in these mice genetically and conditionally induces the expression of caMEK5 in adult neural stem/progenitor cells, enabling the stimulation of adult neurogenesis in the brain. Male ApoE4-KI:caMEK5 and ApoE3-KI:caMEK5 mice were exposed to 0.6 mg/L CdCl2 throughout the experiment, and tamoxifen was administered once Cd-induced impairment in spatial working memory was consistently observed. Cd exposure impaired spatial working memory earlier in ApoE4-KI:caMEK5 than in ApoE3-KI:caMEK5 mice. In both strains, these deficits were rescued after tamoxifen treatment. Consistent with these behavioral findings, tamoxifen treatment enhanced adult neurogenesis by increasing the morphological complexity of adult-born immature neurons. These results provide evidence for a direct link between impaired spatial memory and adult neurogenesis in this GxE model.


Assuntos
Doença de Alzheimer , Apolipoproteína E4 , Camundongos , Animais , Masculino , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Cádmio/metabolismo , Camundongos Transgênicos , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Neurogênese , Transtornos da Memória/metabolismo , Hipocampo/metabolismo , Tamoxifeno/farmacologia , Tamoxifeno/metabolismo , Apolipoproteínas E/metabolismo , Doença de Alzheimer/metabolismo
2.
Drug Metab Dispos ; 50(10): 1414-1428, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35878927

RESUMO

Cadmium (Cd) exposure is associated with increased Alzheimer's disease (AD) risks. The human Apolipoprotein E (ApoE) gene encodes a lipid-transporting protein that is critical for brain functions. Compared with ApoE2 and E3, ApoE4 is associated with increased AD risk. Xenobiotic biotransformation-related genes have been implicated in the pathogenesis of AD. However, little is known about the effects of Cd, ApoE, and sex on drug-processing genes. We investigated the Cd-ApoE interaction on the transcriptomic changes in the brains and livers of ApoE3/ApoE4 transgenic mice. Cd disrupts the transcriptomes of transporter and drug-processing genes in brain and liver in a sex- and ApoE-genotype-specific manner. Proinflammation related genes were enriched in livers of Cd-exposed ApoE4 males, whereas circadian rhythm and lipid metabolism related genes were enriched in livers of Cd-exposed ApoE3 females. In brains, Cd up-regulated the arachidonic acid-metabolizing Cyp2j isoforms only in the brains of ApoE3 mice, whereas the dysregulation of cation transporters was male-specific. In livers, several direct target genes of the major xenobiotic-sensing nuclear receptor pregnane X receptor were uniquely upregulated in Cd-exposed ApoE4 males. There was a female-specific hepatic upregulation of the steroid hormone-metabolizing Cyp2 isoforms and the bile acid synthetic enzyme Cyp7a1 by Cd exposure. The dysregulated liver transporters were mostly involved in intermediary metabolism, with the most significant response observed in ApoE3 females. In conclusion, Cd dysregulated the brain and liver drug-processing genes in a sex- and ApoE-genotype specific manner, and this may serve as a contributing factor for the variance in the susceptibility to Cd neurotoxicity. SIGNIFICANCE STATEMENT: Xenobiotic biotransformation plays an important role in modulating the toxicity of environmental pollutants. The human ApoE4 allele is the strongest genetic risk factor for AD, and cadmium (Cd) is increasingly recognized as an environmental factor of AD. Very little is known regarding the interactions between Cd exposure, sex, and the genes involved in xenobiotic biotransformation in brain and liver. The present study has addressed this critical knowledge gap.


Assuntos
Doença de Alzheimer , Poluentes Ambientais , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/genética , Animais , Apolipoproteína E2/genética , Apolipoproteína E2/metabolismo , Apolipoproteína E2/farmacologia , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Apolipoproteína E3/farmacologia , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Apolipoproteína E4/farmacologia , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Apolipoproteínas E/farmacologia , Ácido Araquidônico/metabolismo , Ácidos e Sais Biliares/metabolismo , Encéfalo/metabolismo , Cádmio/toxicidade , Poluentes Ambientais/metabolismo , Feminino , Predisposição Genética para Doença , Hormônios/metabolismo , Hormônios/farmacologia , Humanos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Receptor de Pregnano X/metabolismo , Isoformas de Proteínas/metabolismo , Xenobióticos/metabolismo
3.
Environ Toxicol ; 37(2): 335-348, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34741586

RESUMO

Cadmium (Cd) is a toxic heavy metal and a significant public health concern. Epidemiological studies suggest that Cd is a potential neurotoxicant, and its exposure is associated with cognitive deficits in children, adults, and seniors. Our previous study has found that adulthood-only Cd exposure can impair cognition in mice. However, few studies have addressed the effects of Cd exposure during adolescence on cognitive behavior in animals later in life. In the present study, we exposed 4-week-old male C57BL/6 mice to 3 mg/L Cd via drinking water for 28 weeks and assessed their hippocampus-dependent learning and memory. Cd did not affect anxiety or locomotor activity in the open field test. However, Cd exposure impaired short-term spatial memory and contextual fear memory in mice. A separate cohort of 4-week-old mice was similarly exposed to Cd for 13 weeks to investigate the potential mechanism of Cd neurotoxicity on cognition. We observed that Cd-treated mice had fewer adult-born cells, adult-born neurons, and a reduced proportion of adult-born cells that differentiated into mature neurons in the subgranular zone of the dentate gyrus. These results suggest that Cd exposure from adolescence to adulthood is sufficient to cause cognitive deficits and impair key processes of hippocampal neurogenesis in mice.


Assuntos
Cádmio , Memória , Animais , Cádmio/toxicidade , Cognição , Hipocampo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurogênese
4.
Proc Natl Acad Sci U S A ; 115(12): E2801-E2810, 2018 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-29507229

RESUMO

Owing to the prevalence of tumor-associated macrophages (TAMs) in cancer and their unique influence upon disease progression and malignancy, macrophage-targeted interventions have attracted notable attention in cancer immunotherapy. However, tractable targets to reduce TAM activities remain very few and far between because the signaling mechanisms underpinning protumor macrophage phenotypes are largely unknown. Here, we have investigated the role of the extracellular-regulated protein kinase 5 (ERK5) as a determinant of macrophage polarity. We report that the growth of carcinoma grafts was halted in myeloid ERK5-deficient mice. Coincidentally, targeting ERK5 in macrophages induced a transcriptional switch in favor of proinflammatory mediators. Further molecular analyses demonstrated that activation of the signal transducer and activator of transcription 3 (STAT3) via Tyr705 phosphorylation was impaired in erk5-deleted TAMs. Our study thus suggests that blocking ERK5 constitutes a treatment strategy to reprogram macrophages toward an antitumor state by inhibiting STAT3-induced gene expression.


Assuntos
Macrófagos/metabolismo , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Fator de Transcrição STAT3/metabolismo , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Polaridade Celular , Humanos , Macrófagos/patologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteína Quinase 7 Ativada por Mitógeno/genética , Fosforilação , Receptores de Superfície Celular/metabolismo , Fator de Transcrição STAT3/genética , Tirosina/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Hum Mol Genet ; 24(10): 2848-60, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25652399

RESUMO

Mitochondrial complex I (NADH dehydrogenase) is a major contributor to neuronal energetics, and mutations in complex I lead to vision loss. Functional, neuroanatomical and transcriptional consequences of complex I deficiency were investigated in retinas of the Ndufs4 knockout mouse. Whole-eye ERGs and multielectrode arrays confirmed a major retinal ganglion cell functional loss at P32, and retinal ganglion cell loss at P42. RNAseq demonstrated a mild and then sharp increase in innate immune and inflammatory retinal transcripts at P22 and P33, respectively, which were confirmed with QRT-PCR. Intraperitoneal injection of the inflammogen lipopolysaccharide further reduced retinal ganglion cell function in Ndufs4 KO, supporting the connection between inflammatory activation and functional loss. Complex I deficiency in the retina clearly caused innate immune and inflammatory markers to increase coincident with loss of vision, and RGC functional loss. How complex I incites inflammation and functional loss is not clear, but could be the result of misfolded complex I generating a 'non-self' response, and induction of innate immune response transcripts was observed before functional loss at P22, including ß-2 microglobulin and Cx3cr1, and during vision loss at P31 (B2m, Tlr 2, 3, 4, C1qa, Cx3cr1 and Fas). These data support the hypothesis that mitochondrial complex I dysfunction in the retina triggers an innate immune and inflammatory response that results in loss of retinal ganglion cell function and death, as in Leber's hereditary Optic Neuropathy and suggests novel therapeutic routes to counter mitochondrial defects that contribute to vision loss.


Assuntos
Complexo I de Transporte de Elétrons/deficiência , Doenças Mitocondriais/fisiopatologia , Retina/fisiopatologia , Células Ganglionares da Retina/fisiologia , Animais , Morte Celular , Complexo I de Transporte de Elétrons/genética , Complexo I de Transporte de Elétrons/imunologia , Feminino , Técnicas de Inativação de Genes , Imunidade Inata/genética , Inflamação/genética , Masculino , Camundongos , Camundongos Knockout , Doenças Mitocondriais/genética , Doenças Mitocondriais/imunologia , Retina/imunologia
6.
J Neurosci ; 35(20): 7833-49, 2015 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-25995470

RESUMO

Recent discoveries have suggested that adult neurogenesis in the subventricular zone (SVZ) and olfactory bulb (OB) may be required for at least some forms of olfactory behavior in mice. However, it is unclear whether conditional and selective enhancement of adult neurogenesis by genetic approaches is sufficient to improve olfactory function under physiological conditions or after injury. Furthermore, specific signaling mechanisms regulating adult neurogenesis in the SVZ/OB are not fully defined. We previously reported that ERK5, a MAP kinase selectively expressed in the neurogenic regions of the adult brain, plays a critical role in adult neurogenesis in the SVZ/OB. Using a site-specific knock-in mouse model, we report here that inducible and targeted activation of the endogenous ERK5 in adult neural stem/progenitor cells enhances adult neurogenesis in the OB by increasing cell survival and neuronal differentiation. This conditional ERK5 activation also improves short-term olfactory memory and odor-cued associative olfactory learning under normal physiological conditions. Furthermore, these mice show enhanced recovery of olfactory function and have more adult-born neurons after a zinc sulfate-induced lesion of the main olfactory epithelium. We conclude that ERK5 MAP kinase is an important endogenous signaling pathway regulating adult neurogenesis in the SVZ/OB, and that conditional activation of endogenous ERK5 is sufficient to enhance adult neurogenesis in the OB thereby improving olfactory function both under normal conditions and after injury.


Assuntos
Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Neurogênese , Neurônios/metabolismo , Bulbo Olfatório/metabolismo , Olfato , Animais , Células Cultivadas , Memória , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 7 Ativada por Mitógeno/genética , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/fisiologia , Neurônios/citologia , Neurônios/fisiologia , Bulbo Olfatório/citologia , Bulbo Olfatório/crescimento & desenvolvimento , Bulbo Olfatório/fisiologia , Transdução de Sinais
7.
J Neurosci ; 34(6): 2130-47, 2014 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-24501354

RESUMO

Recent studies have shown that inhibition of adult neurogenesis impairs the formation of hippocampus-dependent memory. However, it is not known whether increasing adult neurogenesis affects the persistence of hippocampus-dependent long-term memory. Furthermore, signaling mechanisms that regulate adult neurogenesis are not fully defined. We recently reported that the conditional and targeted knock-out of ERK5 MAP kinase in adult neurogenic regions of the mouse brain attenuates adult neurogenesis in the hippocampus and disrupts several forms of hippocampus-dependent memory. Here, we developed a gain-of-function knock-in mouse model to specifically activate endogenous ERK5 in the neurogenic regions of the adult brain. We report that the selective and targeted activation of ERK5 increases adult neurogenesis in the dentate gyrus by enhancing cell survival, neuronal differentiation, and dendritic complexity. Conditional ERK5 activation also improves the performance of challenging forms of spatial learning and memory and extends hippocampus-dependent long-term memory. We conclude that enhancing signal transduction of a single signaling pathway within adult neural stem/progenitor cells is sufficient to increase adult neurogenesis and improve the persistence of hippocampus-dependent memory. Furthermore, activation of ERK5 may provide a novel therapeutic target to improve long-term memory.


Assuntos
Hipocampo/enzimologia , Memória de Longo Prazo/fisiologia , Proteína Quinase 7 Ativada por Mitógeno/genética , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Neurogênese/fisiologia , Fatores Etários , Animais , Animais Recém-Nascidos , Diferenciação Celular/fisiologia , Ativação Enzimática/fisiologia , Técnicas de Introdução de Genes , Hipocampo/citologia , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
8.
J Biol Chem ; 288(4): 2623-31, 2013 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-23223235

RESUMO

Prolactin-stimulated adult neurogenesis in the subventricular zone (SVZ) and olfactory bulb (OB) mediates several reproductive behaviors including mating/pregnancy, dominant male pheromone preference in females, and paternal recognition of offspring. However, downstream signaling mechanisms underlying prolactin-induced adult neurogenesis are completely unknown. We report here for the first time that prolactin activates extracellular signal-regulated kinase 5 (ERK5), a MAP kinase that is specifically expressed in the neurogenic regions of the adult mouse brain. Knockdown of ERK5 by retroviral infection of shRNA attenuates prolactin-stimulated neurogenesis in SVZ-derived adult neural stem/progenitor cells (aNPCs). Inducible erk5 deletion in adult neural stem cells of transgenic mice inhibits neurogenesis in the SVZ and OB following prolactin infusion or mating/pregnancy. These results identify ERK5 as a novel and critical signaling mechanism underlying prolactin-induced adult neurogenesis.


Assuntos
Encéfalo/metabolismo , Proteína Quinase 7 Ativada por Mitógeno/fisiologia , Bulbo Olfatório/metabolismo , Prolactina/metabolismo , Animais , Mapeamento Encefálico/métodos , Feminino , Deleção de Genes , Genótipo , Camundongos , Camundongos Knockout , Microscopia Confocal/métodos , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Neurogênese , Proteínas Recombinantes/metabolismo , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais , Tamoxifeno/farmacologia
9.
Toxicol Sci ; 200(1): 199-212, 2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-38579196

RESUMO

Cadmium (Cd) is a ubiquitous toxic heavy metal and a potential neurotoxicant due to its wide use in industrial manufacturing processes and commercial products, including fertilizers. The general population is exposed to Cd through food and smoking due to high transfer rates of Cd from contaminated soil. Cd has been shown to mimic calcium ions (Ca2+) and interfere with intracellular Ca2+ levels and Ca2+ signaling in in vitro studies. However, nothing is known about Cd's effects on Ca2+ activity in neurons in live animals. This study aimed to determine if Cd disrupts Ca2+ transients of neurons in CA1 region of the hippocampus during an associative learning paradigm. We utilized in vivo Ca2+ imaging in awake, freely moving C57BL/6 mice to measure Ca2+ activity in CA1 excitatory neurons expressing genetically encoded Ca2+ sensor GCaMP6 during an associative learning paradigm. We found that a smaller proportion of neurons are activated in Cd-treated groups compared with control during fear conditioning, suggesting that Cd may contribute to learning and memory deficit by reducing the activity of neurons. We observed these effects at Cd exposure levels that result in blood Cd levels comparable with the general U.S. population levels. This provides a possible molecular mechanism for Cd interference of learning and memory at exposure levels relevant to U.S. adults. To our knowledge, our study is the first to describe Cd effects on brain Ca2+ activity in vivo in freely behaving mice. This study provides evidence for impairment of neuronal calcium activity in hippocampal CA1 excitatory neurons in freely moving mice following cadmium exposure.


Assuntos
Região CA1 Hipocampal , Camundongos Endogâmicos C57BL , Animais , Região CA1 Hipocampal/efeitos dos fármacos , Região CA1 Hipocampal/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Cálcio/metabolismo , Masculino , Cádmio/toxicidade , Camundongos , Sinalização do Cálcio/efeitos dos fármacos , Medo/efeitos dos fármacos , Cloreto de Cádmio/toxicidade
10.
J Neurosci ; 32(45): 15769-78, 2012 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-23136416

RESUMO

Cilia of olfactory sensory neurons are the primary sensory organelles for olfaction. The detection of odorants by the main olfactory epithelium (MOE) depends on coupling of odorant receptors to the type 3 adenylyl cyclase (AC3) in olfactory cilia. We monitored the effect of airflow on electro-olfactogram (EOG) responses and found that the MOE of mice can sense mechanical forces generated by airflow. The airflow-sensitive EOG response in the MOE was attenuated when cAMP was increased by odorants or by forskolin suggesting a common mechanism for airflow and odorant detection. In addition, the sensitivity to airflow was significantly impaired in the MOE from AC3(-/-) mice. We conclude that AC3 in the MOE is required for detecting the mechanical force of airflow, which in turn may regulate odorant perception during sniffing.


Assuntos
Adenilil Ciclases/metabolismo , Mucosa Olfatória/metabolismo , Percepção Olfatória/fisiologia , Olfato/fisiologia , Adenilil Ciclases/genética , Animais , Feminino , Masculino , Camundongos , Camundongos Knockout , Odorantes , Mucosa Olfatória/efeitos dos fármacos , Percepção Olfatória/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Olfato/efeitos dos fármacos
11.
J Neurosci ; 32(19): 6444-55, 2012 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-22573667

RESUMO

Although there is evidence suggesting that adult neurogenesis may contribute to hippocampus-dependent memory, signaling mechanisms responsible for adult hippocampal neurogenesis are not well characterized. Here we report that ERK5 mitogen-activated protein kinase is specifically expressed in the neurogenic regions of the adult mouse brain. The inducible and conditional knock-out (icKO) of erk5 specifically in neural progenitors of the adult mouse brain attenuated adult hippocampal neurogenesis. It also caused deficits in several forms of hippocampus-dependent memory, including contextual fear conditioning generated by a weak footshock. The ERK5 icKO mice were also deficient in contextual fear extinction and reversal of Morris water maze spatial learning and memory, suggesting that adult neurogenesis plays an important role in hippocampus-dependent learning flexibility. Furthermore, our data suggest a critical role for ERK5-mediated adult neurogenesis in pattern separation, a form of dentate gyrus-dependent spatial learning and memory. Moreover, ERK5 icKO mice have no memory 21 d after training in the passive avoidance test, suggesting a pivotal role for adult hippocampal neurogenesis in the expression of remote memory. Together, our results implicate ERK5 as a novel signaling molecule regulating adult neurogenesis and provide strong evidence that adult neurogenesis is critical for several forms of hippocampus-dependent memory formation, including fear extinction, and for the expression of remote memory.


Assuntos
Extinção Psicológica/fisiologia , Medo/fisiologia , Deleção de Genes , Memória de Longo Prazo/fisiologia , Proteína Quinase 7 Ativada por Mitógeno/deficiência , Proteína Quinase 7 Ativada por Mitógeno/genética , Inibição Neural/genética , Neurogênese/fisiologia , Envelhecimento/genética , Animais , Giro Denteado/enzimologia , Giro Denteado/fisiologia , Marcação de Genes/métodos , Masculino , Camundongos , Camundongos Knockout , Neurogênese/genética , Distribuição Aleatória , Transdução de Sinais/genética
12.
J Neurosci ; 32(12): 4118-32, 2012 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-22442076

RESUMO

ERK5 MAP kinase is highly expressed in the developing nervous system and has been implicated in promoting the survival of immature neurons in culture. However, its role in the development and function of the mammalian nervous system has not been established in vivo. Here, we report that conditional deletion of the erk5 gene in mouse neural stem cells during development reduces the number of GABAergic interneurons in the main olfactory bulb (OB). Our data suggest that this is due to a decrease in proliferation and an increase in apoptosis in the subventricular zone and rostral migratory stream of ERK5 mutant mice. Interestingly, ERK5 mutant mice have smaller OB and are impaired in odor discrimination between structurally similar odorants. We conclude that ERK5 is a novel signaling pathway regulating developmental OB neurogenesis and olfactory behavior.


Assuntos
Neurônios GABAérgicos/fisiologia , Proteína Quinase 7 Ativada por Mitógeno/deficiência , Odorantes , Bulbo Olfatório , Transtornos da Percepção/genética , Transtornos da Percepção/patologia , Fatores Etários , Análise de Variância , Animais , Animais Recém-Nascidos , Apoptose/genética , Bromodesoxiuridina/metabolismo , Movimento Celular , Modelos Animais de Doenças , Eletroculografia/métodos , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento/genética , Glutamato Descarboxilase/metabolismo , Marcação In Situ das Extremidades Cortadas , Ventrículos Laterais/embriologia , Ventrículos Laterais/crescimento & desenvolvimento , Ventrículos Laterais/patologia , Camundongos , Camundongos Transgênicos , Proteína Quinase 7 Ativada por Mitógeno/genética , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Células-Tronco Neurais/fisiologia , Neurogênese/genética , Bulbo Olfatório/embriologia , Bulbo Olfatório/crescimento & desenvolvimento , Bulbo Olfatório/patologia , Fosfopiruvato Hidratase/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Ácidos Siálicos/metabolismo , Transdução de Sinais , Olfato/genética
13.
J Biol Chem ; 287(28): 23306-17, 2012 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-22645146

RESUMO

Recent studies have led to the exciting idea that adult-born neurons in the dentate gyrus of the hippocampus may play a role in hippocampus-dependent memory formation. However, signaling mechanisms that regulate adult hippocampal neurogenesis are not well defined. Here we report that extracellular signal-regulated kinase 5 (ERK5), a member of the mitogen-activated protein kinase family, is selectively expressed in the neurogenic regions of the adult mouse brain. We present evidence that shRNA suppression of ERK5 in adult hippocampal neural stem/progenitor cells (aNPCs) reduces the number of neurons while increasing the number of cells expressing markers for stem/progenitor cells or proliferation. Furthermore, shERK5 attenuates both transcription and neuronal differentiation mediated by Neurogenin 2, a transcription factor expressed in adult hippocampal neural progenitor cells. By contrast, ectopic activation of endogenous ERK5 signaling via expression of constitutive active MEK5, an upstream activating kinase for ERK5, promotes neurogenesis in cultured aNPCs and in the dentate gyrus of the mouse brain. Moreover, neurotrophins including NT3 activate ERK5 and stimulate neuronal differentiation in aNPCs in an ERK5-dependent manner. Finally, inducible and conditional deletion of ERK5 specifically in the neurogenic regions of the adult mouse brain delays the normal progression of neuronal differentiation and attenuates adult neurogenesis in vivo. These data suggest ERK5 signaling as a critical regulator of adult hippocampal neurogenesis.


Assuntos
Hipocampo/metabolismo , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese , Animais , Antineoplásicos Hormonais/farmacologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Western Blotting , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Giro Denteado/citologia , Giro Denteado/crescimento & desenvolvimento , Giro Denteado/metabolismo , Hipocampo/citologia , Hipocampo/crescimento & desenvolvimento , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , MAP Quinase Quinase 5/genética , MAP Quinase Quinase 5/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia Confocal , Proteína Quinase 7 Ativada por Mitógeno/genética , Células NIH 3T3 , Fatores de Crescimento Neural/farmacologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/efeitos dos fármacos , Interferência de RNA , Tamoxifeno/farmacologia
14.
Neurobiol Learn Mem ; 105: 81-92, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23871742

RESUMO

Adult neurogenesis occurs in two discrete regions of the adult mammalian brain, the subgranular zone (SGZ) of the dentate gyrus (DG) and the subventricular zone (SVZ) along the lateral ventricles. Signaling mechanisms regulating adult neurogenesis in the SGZ are currently an active area of investigation. Adult-born neurons in the DG functionally integrate into the hippocampal circuitry and form functional synapses, suggesting a role for these neurons in hippocampus-dependent memory formation. Although results from earlier behavioral studies addressing this issue were inconsistent, recent advances in conditional gene targeting technology, viral injection and optogenetic approaches have provided convincing evidence supporting a role for adult-born neurons in the more challenging forms of hippocampus-dependent learning and memory. Here, we briefly summarize these recent studies with a focus on extra signal-regulated kinase (ERK) 5, a MAP kinase whose expression in the adult brain is restricted to the neurogenic regions including the SGZ and SVZ. We review evidence identifying ERK5 as a novel endogenous signaling pathway that regulates the pro-neural transcription factor Neurogenin 2, is activated by neurotrophins and is critical for adult neurogenesis. We discuss studies demonstrating that specific deletion of ERK5 in the adult neurogenic regions impairs several forms of hippocampus-dependent memory formation in mice. These include contextual fear memory extinction, the establishment and maintenance of remote contextual fear memory, and several other challenging forms of hippocampus-dependent memory formation including 48h memory for novel object recognition, contextual fear memory established by a weak foot shock, pattern separation, and reversal of spatial learning and memory. We also briefly discuss current evidence that increasing adult neurogenesis, by small molecules or genetic manipulation, improves memory formation and long-term memory.


Assuntos
Giro Denteado/fisiologia , Extinção Psicológica/fisiologia , Memória/fisiologia , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Neurogênese , Animais , Giro Denteado/citologia , Giro Denteado/enzimologia , Medo/fisiologia , Camundongos , Camundongos Knockout , Neurogênese/genética
15.
Learn Mem ; 19(9): 369-74, 2012 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-22904367

RESUMO

One of the intriguing questions in neurobiology is how long-term memory (LTM) traces are established and maintained in the brain. Memory can be divided into at least two temporally and mechanistically distinct forms. Short-term memory (STM) lasts no longer than several hours, while LTM persists for days or longer. A crucial step in the generation of LTM is consolidation, a process in which STM is converted to LTM. Hippocampus-dependent LTM depends on activation of Ca(2+), Erk/MAP kinase (MAPK), and cAMP signaling pathways, as well as de novo gene expression and translation. One of the transcriptional pathways strongly implicated in LTM is the CREB/CRE (calcium, cAMP response element) transcriptional pathway. Interestingly, this transcriptional pathway may also contribute to other forms of neuroplasticity including adaptive responses to drugs. Evidence discussed in this review indicates that activation of the Erk1/2 MAP Kinase (MAPK)/CRE transcriptional pathway during the formation of hippocampus-dependent memory depends on calmodulin (CaM)-stimulated adenylyl cyclases.


Assuntos
Adenilil Ciclases/metabolismo , Hipocampo/enzimologia , Memória/fisiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Transdução de Sinais/fisiologia , Animais , Cálcio/metabolismo , Hipocampo/citologia , Humanos , Modelos Biológicos , Plasticidade Neuronal/fisiologia
16.
J Cell Biol ; 177(2): 253-64, 2007 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-17452529

RESUMO

Elucidation of mechanisms regulating cell cycle progression is of fundamental importance for cell and cancer biology. Although several genes and signaling pathways are implicated in G1-S regulation, less is known regarding the mechanisms controlling cell cycle progression through G2 and M phases. We report that extracellular signal-regulated kinase 5 (ERK5), a member of the mitogen-activated protein kinases, is activated at G2-M and required for timely mitotic entry. Stimulation of ERK5 activated nuclear factor kappaB (NFkappaB) through ribosomal S6 kinase 2 (RSK2)-mediated phosphorylation and degradation of IkappaB. Furthermore, selective inhibition of NFkappaB at G2-M phases substantially delayed mitotic entry and inhibited transcription of G2-M-specific genes, including cyclin B1, cyclin B2, Plk-1, and cdc25B. Moreover, inhibition of NFkappaB at G2-M diminished mitosis induced by constitutive activation of ERK5, providing a direct link between ERK5, NFkappaB, and regulation of G2-M progression. We conclude that a novel ERK5-NFkappaB signaling pathway plays a key role in regulation of the G2-M progression.


Assuntos
Fase G2 , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Mitose , NF-kappa B/metabolismo , Linhagem Celular , Células HeLa , Humanos , Proteínas I-kappa B/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Transdução de Sinais
17.
Proc Natl Acad Sci U S A ; 105(39): 15136-41, 2008 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-18812510

RESUMO

Inhibition of mitochondrial complex I is one of the leading hypotheses for dopaminergic neuron death associated with Parkinson's disease (PD). To test this hypothesis genetically, we used a mouse strain lacking functional Ndufs4, a gene encoding a subunit required for complete assembly and function of complex I. Deletion of the Ndufs4 gene abolished complex I activity in midbrain mesencephalic neurons cultured from embryonic day (E) 14 mice, but did not affect the survival of dopaminergic neurons in culture. Although dopaminergic neurons were more sensitive than other neurons in these cultures to cell death induced by rotenone, MPP(+), or paraquat treatments, the absence of complex I activity did not protect the dopaminergic neurons, as would be expected if these compounds act by inhibiting complex 1. In fact, the dopaminergic neurons were more sensitive to rotenone. These data suggest that dopaminergic neuron death induced by treatment with rotenone, MPP(+), or paraquat is independent of complex I inhibition.


Assuntos
Apoptose , Dopamina/metabolismo , NADH Desidrogenase/antagonistas & inibidores , Neurônios/patologia , Doença de Parkinson/patologia , Animais , Deleção de Genes , Camundongos , Camundongos Mutantes , NADH Desidrogenase/genética , NADH Desidrogenase/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Paraquat/toxicidade , Doença de Parkinson/enzimologia , Rotenona/toxicidade
18.
Commun Biol ; 4(1): 1398, 2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34912029

RESUMO

The human Apolipoprotein E4 (ApoE4) variant is the strongest known genetic risk factor for Alzheimer's disease (AD). Cadmium (Cd) has been shown to impair learning and memory at a greater extent in humanized ApoE4 knock-in (ApoE4-KI) mice as compared to ApoE3 (common allele)-KI mice. Here, we determined how cadmium interacts with ApoE4 gene variants to modify the gut-liver axis. Large intestinal content bacterial 16S rDNA sequencing, serum lipid metabolomics, and hepatic transcriptomics were analyzed in ApoE3- and ApoE4-KI mice orally exposed to vehicle, a low dose, or a high dose of Cd in drinking water. ApoE4-KI males had the most prominent changes in their gut microbiota, as well as a predicted down-regulation of many essential microbial pathways involved in nutrient and energy homeostasis. In the host liver, cadmium-exposed ApoE4-KI males had the most differentially regulated pathways; specifically, there was enrichment in several pathways involved in platelet activation and drug metabolism. In conclusion, Cd exposure profoundly modified the gut-liver axis in the most susceptible mouse strain to neurological damage namely the ApoE4-KI males, evidenced by an increase in microbial AD biomarkers, reduction in energy supply-related pathways in gut and blood, and an increase in hepatic pathways involved in inflammation and xenobiotic biotransformation.


Assuntos
Doença de Alzheimer/metabolismo , Cádmio/metabolismo , Doença de Alzheimer/fisiopatologia , Animais , Modelos Animais de Doenças , Feminino , Intestino Grosso/metabolismo , Fígado/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Vacina contra Caxumba
19.
Neurotoxicology ; 81: 127-136, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33039505

RESUMO

Cadmium (Cd) is a heavy metal that is one of the most toxic environmental pollutants throughout the world. We previously reported that Cd exposure impairs olfactory memory in mice. However, the underlying mechanisms for its neurotoxicity for olfactory function are not well understood. Since adult Subventricular zone (SVZ) and Olfactory Bulb (OB) neurogenesis contributes to olfaction, olfactory memory defects caused by Cd may be due to inhibition of neurogenesis. In this study, using bromodeoxyuridine (BrdU) labeling and immunohistochemistry, we found that 0.6 mg/L Cd exposure through drinking water impaired adult SVZ/OB neurogenesis in C57BL/6 mice. To determine if the inhibition of olfactory memory by Cd can be reversed by stimulating adult neurogenesis, we utilized the transgenic caMEK5 mouse strain to conditional stimulate of adult neurogenesis by activating the endogenous ERK5 MAP kinase signaling pathway. This was accomplished by conditionally induced expression of active MEK5 (caMEK5) in adult neural stem/progenitor cells. The caMEK5 mice were exposed to 0.6 mg/L Cd for 38 weeks, and tamoxifen was administered to induce caMEK5 expression and stimulate adult SVZ/OB neurogenesis during Cd exposure. Short-term olfactory memory test and sand-digging based, odor-cued olfactory learning and memory test were conducted after Cd and tamoxifen treatments to examine their effects on olfaction. Here we report that Cd exposure impaired short-term olfactory memory and odor-cued associative learning and memory in mice. Furthermore, the Cd-impaired olfactory memory deficits were rescued by the tamoxifen-induction of caMEK5 expression. This suggests that Cd exposure impairs olfactory function by affecting adult SVZ/OB neurogenesis in mice.


Assuntos
Comportamento Animal , Ventrículos Laterais/enzimologia , Memória , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Neurogênese , Transtornos do Olfato/prevenção & controle , Bulbo Olfatório/enzimologia , Percepção Olfatória , Olfato , Animais , Aprendizagem por Associação , Cloreto de Cádmio , Sinais (Psicologia) , Modelos Animais de Doenças , Ativação Enzimática , Feminino , Ventrículos Laterais/patologia , Ventrículos Laterais/fisiopatologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteína Quinase 7 Ativada por Mitógeno/genética , Odorantes , Transtornos do Olfato/induzido quimicamente , Transtornos do Olfato/enzimologia , Transtornos do Olfato/fisiopatologia , Bulbo Olfatório/patologia , Bulbo Olfatório/fisiopatologia , Fatores de Tempo
20.
Toxicol Sci ; 173(1): 189-201, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31626305

RESUMO

Cadmium (Cd) is a heavy metal of great public health concern. Recent studies suggested a link between Cd exposure and cognitive decline in humans. The ε4 allele, compared with the common ε3 allele, of the human apolipoprotein E gene (ApoE) is associated with accelerated cognitive decline and increased risks for Alzheimer's disease (AD). To investigate the gene-environment interactions (GxE) between ApoE-ε4 and Cd exposure on cognition, we used a mouse model of AD that expresses human ApoE-ε3 (ApoE3-KI [knock-in]) or ApoE-ε4 (ApoE4-KI). Mice were exposed to 0.6 mg/l CdCl2 through drinking water for 14 weeks and assessed for hippocampus-dependent memory. A separate cohort was sacrificed immediately after exposure and used for Cd measurements and immunostaining. The peak blood Cd was 0.3-0.4 µg/l, within levels found in the U.S. general population. All Cd-treated animals exhibited spatial working memory deficits in the novel object location test. This deficit manifested earlier in ApoE4-KI mice than in ApoE3-KI within the same sex and earlier in males than females within the same genotype. ApoE4-KI but not ApoE3-KI mice exhibited reduced spontaneous alternation later in life in the T-maze test. Finally, Cd exposure impaired neuronal differentiation of adult-born neurons in the hippocampus of male ApoE4-KI mice. These data suggest that a GxE between ApoE4 and Cd exposure leads to accelerated cognitive impairment and that impaired adult hippocampal neurogenesis may be one of the underlying mechanisms. Furthermore, male mice were more susceptible than female mice to this GxE effect when animals were young.


Assuntos
Apolipoproteína E4/metabolismo , Cádmio/toxicidade , Substâncias Perigosas/toxicidade , Memória/efeitos dos fármacos , Doença de Alzheimer , Animais , Apolipoproteína E3 , Comportamento Animal , Encéfalo , Cognição , Modelos Animais de Doenças , Feminino , Interação Gene-Ambiente , Hipocampo , Humanos , Masculino , Aprendizagem em Labirinto , Transtornos da Memória , Camundongos , Camundongos Transgênicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA