Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Exp Cell Res ; 368(1): 84-100, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29689278

RESUMO

Development of the spinal cord requires dynamic and tightly controlled expression of numerous transcription factors. Forkhead Box protein J1 (FoxJ1) is a transcription factor involved in ciliogenesis and is specifically expressed in ependymal cells (ECs) in the adult central nervous system. However, using FoxJ1 fate-mapping mouse lines, we observed that FoxJ1 is also transiently expressed by the progenitors of other neural subtypes during development. Moreover, using a knock-in mouse line, we discovered that FoxJ1 is essential for embryonic progenitors to follow a normal developmental trajectory. FoxJ1 loss perturbed embryonic progenitor proliferation and cell fate determination, and resulted in formation of adult ECs having impaired stem cell potential and an inability to respond to spinal cord injury in both male and female animals. Thus, our study uncovers unexpected developmental functions of FoxJ1 in cell fate determination of subsets of neural cells and suggests that FoxJ1 is critical for maintaining the stem cell potential of ECs into adulthood.


Assuntos
Diferenciação Celular/fisiologia , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica/genética , Células-Tronco/citologia , Animais , Epêndima/metabolismo , Feminino , Masculino , Camundongos , Organogênese/fisiologia , Medula Espinal/metabolismo , Traumatismos da Medula Espinal/metabolismo
2.
Sci Rep ; 14(1): 7742, 2024 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-38565895

RESUMO

Evidence from genetic and epidemiological studies point to lipid metabolism defects in both the brain and periphery being at the core of Alzheimer's disease (AD) pathogenesis. Previously, we reported that central inhibition of the rate-limiting enzyme in monounsaturated fatty acid synthesis, stearoyl-CoA desaturase (SCD), improves brain structure and function in the 3xTg mouse model of AD (3xTg-AD). Here, we tested whether these beneficial central effects involve recovery of peripheral metabolic defects, such as fat accumulation and glucose and insulin handling. As early as 3 months of age, 3xTg-AD mice exhibited peripheral phenotypes including increased body weight and visceral and subcutaneous white adipose tissue as well as diabetic-like peripheral gluco-regulatory abnormalities. We found that intracerebral infusion of an SCD inhibitor that normalizes brain fatty acid desaturation, synapse loss and learning and memory deficits in middle-aged memory-impaired 3xTg-AD mice did not affect these peripheral phenotypes. This suggests that the beneficial effects of central SCD inhibition on cognitive function are not mediated by recovery of peripheral metabolic abnormalities. Given the widespread side-effects of systemically administered SCD inhibitors, these data suggest that selective inhibition of SCD in the brain may represent a clinically safer and more effective strategy for AD.


Assuntos
Doença de Alzheimer , Estearoil-CoA Dessaturase , Camundongos , Animais , Estearoil-CoA Dessaturase/genética , Estearoil-CoA Dessaturase/metabolismo , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Metabolismo dos Lipídeos/fisiologia , Lipogênese , Modelos Animais de Doenças , Camundongos Transgênicos
3.
J Neurosci ; 32(43): 15012-26, 2012 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-23100423

RESUMO

Adult forebrain neurogenesis is dynamically regulated. Multiple families of niche-derived cues have been implicated in this regulation, but the precise roles of key intracellular signaling pathways remain vaguely defined. Here, we show that mammalian target of rapamycin (mTOR) signaling is pivotal in determining proliferation versus quiescence in the adult forebrain neural stem cell (NSC) niche. Within this niche, mTOR complex-1 (mTORC1) activation displays stage specificity, occurring in transiently amplifying (TA) progenitor cells but not in GFAP+ stem cells. Inhibiting mTORC1 depletes the TA progenitor pool in vivo and suppresses epidermal growth factor (EGF)-induced proliferation within neurosphere cultures. Interestingly, mTORC1 inhibition induces a quiescence-like phenotype that is reversible. Likewise, mTORC1 activity and progenitor proliferation decline within the quiescent NSC niche of the aging brain, while EGF administration reactivates the quiescent niche in an mTORC1-dependent manner. These findings establish fundamental links between mTOR signaling, proliferation, and aging-associated quiescence in the adult forebrain NSC niche.


Assuntos
Envelhecimento , Diferenciação Celular/fisiologia , Células-Tronco Neurais/fisiologia , Prosencéfalo/citologia , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR/metabolismo , 2',3'-Nucleotídeo Cíclico Fosfodiesterases/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Cultivadas , Proteínas do Domínio Duplacortina , Embrião de Mamíferos , Feminino , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fatores de Crescimento de Fibroblastos/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteína Glial Fibrilar Ácida/genética , Proteínas de Fluorescência Verde/genética , Humanos , Antígeno Ki-67/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microdissecção , Proteínas Associadas aos Microtúbulos/metabolismo , Fatores de Crescimento Neural/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/efeitos dos fármacos , Neuropeptídeos/metabolismo , Fator de Transcrição 2 de Oligodendrócitos , Gravidez , Proteína S6 Ribossômica/metabolismo , Subunidade beta da Proteína Ligante de Cálcio S100 , Proteínas S100/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Sirolimo/metabolismo , Serina-Treonina Quinases TOR/genética , Transfecção , Tubulina (Proteína)/metabolismo
4.
Genes Chromosomes Cancer ; 51(8): 792-804, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22514116

RESUMO

Neuroblastoma (NB) is the most common and lethal extracranial solid tumor of childhood. Despite aggressive therapy, more than half of the children with advanced NB will die of uncontrolled metastatic disease. After chemotherapy, tumor-initiating cells (TICs) could persist, cause relapses and metastasis. The aim of this study is to demonstrate the tumor-initiating properties of CD133high NB cells and to identify new specific genetic abnormalities. Isolation of the CD133high cell population from NB cell lines was followed by neurosphere formation, soft agar assays, and orthotopic injections in NOD/SCID/IL2Rγc-null mice. A differential genotyping analysis was performed with Affymetrix SNP 6.0 arrays on CD133low and CD133high populations and the frequency of the abnormalities of 36 NB tumors was determined. Our results show that CD133high NB cells possess tumor-initiating properties, as CD133high cells formed significantly more neurospheres and produced significantly more colonies in soft agar than CD133low. Injection of 500 CD133high cells was sufficient to generate primary tumors and frequent metastases in mice. Differential genotyping analysis demonstrated two common regions with gains (16p13.3 and 19p13.3) including the gene EFNA2 in the CD133high population, and two with loss of heterozygosity (16q12.1 and 21q21.3) in the CD133low population. The gain of EFNA2 correlated with increased expression of the corresponding protein. These abnormalities were found in NB samples and some were significantly correlated with CD133 expression. Our results show that CD133high NB cells have TICs properties and present different genotyping characteristics compared to CD133low cells. Our findings reveal insights into new therapeutic targets in NB TICs.


Assuntos
Antígenos CD/genética , Glicoproteínas/genética , Neuroblastoma/genética , Peptídeos/genética , Antígeno AC133 , Neoplasias das Glândulas Suprarrenais , Animais , Antígenos CD/biossíntese , Antígenos CD/metabolismo , Linhagem Celular Tumoral , Separação Celular , Distribuição de Qui-Quadrado , Aberrações Cromossômicas , Feminino , Genótipo , Glicoproteínas/biossíntese , Glicoproteínas/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Mutagênese Insercional , Transplante de Neoplasias , Neuroblastoma/metabolismo , Peptídeos/metabolismo , Polimorfismo de Nucleotídeo Único
5.
Nat Commun ; 13(1): 2061, 2022 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-35443751

RESUMO

The defining features of Alzheimer's disease (AD) include alterations in protein aggregation, immunity, lipid metabolism, synapses, and learning and memory. Of these, lipid abnormalities are the least understood. Here, we investigate the role of Stearoyl-CoA desaturase (SCD), a crucial regulator of fatty acid desaturation, in AD pathogenesis. We show that inhibiting brain SCD activity for 1-month in the 3xTg mouse model of AD alters core AD-related transcriptomic pathways in the hippocampus, and that it concomitantly restores essential components of hippocampal function, including dendritic spines and structure, immediate-early gene expression, and learning and memory itself. Moreover, SCD inhibition dampens activation of microglia, key mediators of spine loss during AD and the main immune cells of the brain. These data reveal that brain fatty acid metabolism links AD genes to downstream immune, synaptic, and functional impairments, identifying SCD as a potential target for AD treatment.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Doença de Alzheimer/metabolismo , Animais , Disfunção Cognitiva/metabolismo , Modelos Animais de Doenças , Ácidos Graxos/metabolismo , Hipocampo/metabolismo , Camundongos , Camundongos Transgênicos , Estearoil-CoA Dessaturase/genética , Estearoil-CoA Dessaturase/metabolismo
6.
Hippocampus ; 21(12): 1334-47, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20623741

RESUMO

Hippocampal neurogenesis continues into adulthood in mammalian vertebrates, and in experimental rodent models it is powerfully stimulated by exposure to a voluntary running wheel. In this study, we demonstrate that exposure to a running wheel environment, in the absence of running, is sufficient to regulate specific aspects of hippocampal neurogenesis. Adult mice were provided with standard housing, housing enriched with a running wheel or housing enriched with a locked wheel (i.e., an environment comparable to that of running animals, without the possibility of engaging in running). We found that mice in the running wheel and locked wheel groups exhibited equivalent increases in proliferation within the neurogenic niche of the dentate gyrus; this included comparable increases in the proliferation of radial glia-like stem cells and the number of proliferating neuroblasts. However, only running animals displayed increased numbers of postmitotic neuroblasts and mature neurons. These results demonstrate that the running wheel environment itself is sufficient for promoting proliferation of early lineage hippocampal precursors, while running per se enables newly generated neuroblasts to survive and mature into functional hippocampal neurons. Thus, both running-independent and running-dependent stimuli are integral to running wheel-induced hippocampal neurogenesis.


Assuntos
Giro Denteado/citologia , Abrigo para Animais , Neurogênese/fisiologia , Neurônios/citologia , Corrida/fisiologia , Equipamentos Esportivos , Animais , Contagem de Células , Divisão Celular , Giro Denteado/fisiologia , Masculino , Camundongos , Células-Tronco/citologia
7.
Front Neurosci ; 15: 621076, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33841077

RESUMO

The ventricular-subventricular zone (V-SVZ) is the principal neurogenic niche in the adult mammalian forebrain. Neural stem/progenitor cell (NSPC) activity within the V-SVZ is controlled by numerous of extrinsic factors, whose downstream effects on NSPC proliferation, survival and differentiation are transduced via a limited number of intracellular signaling pathways. Here, we investigated the relationship between age-related changes in NSPC output and activity of signaling pathways downstream of the epidermal growth factor receptor (EGFR), a major regulator of NSPC activity. Biochemical experiments indicated that age-related decline of NSPC activity in vivo is accompanied by selective deficits amongst various EGFR-induced signal pathways within the V-SVZ niche. Pharmacological loss-of-function signaling experiments with cultured NSPCs revealed both overlap and selectivity in the biological functions modulated by the EGFR-induced PI3K/AKT, MEK/ERK and mTOR signaling modules. Specifically, while all three modules promoted EGFR-mediated NSPC proliferation, only mTOR contributed to NSPC survival and only MEK/ERK repressed NSPC differentiation. Using a gain-of-function in vivo genetic approach, we electroporated a constitutively active EGFR construct into a subpopulation of quiescent, EGFR-negative neural stem cells (qNSCs); this ectopic activation of EGFR signaling enabled qNSCs to divide in 3-month-old early adult mice, but not in mice at middle-age or carrying familial Alzheimer disease mutations. Thus, (i) individual EGFR-induced signaling pathways have dissociable effects on NSPC proliferation, survival, and differentiation, (ii) activation of EGFR signaling is sufficient to stimulate qNSC cell cycle entry during early adulthood, and (iii) the proliferative effects of EGFR-induced signaling are dominantly overridden by anti-proliferative signals associated with aging and Alzheimer's disease.

8.
Reprod Sci ; 28(7): 2006-2011, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33405213

RESUMO

Preterm birth is the first cause of neonatal mortality and is associated with elevated risks of long-term complications such as neurodevelopmental impairment. Prediction of spontaneous preterm birth, one of the biggest challenges in obstetrics, aims at delaying birth in order to allow corticosteroid therapy and, if necessary, transfer of patient to a higher-level maternity care unit. We aimed to assess the predictive role of phIGFBP-1 (Actim® Partus) diagnostic test on patients at risk of preterm labor, routinely used in our institution. We conducted a retrospective cohort study on 99 patients admitted in the high-risk pregnancy unit of our institution from June 2012 to November 2014. The primary outcome measures were delivery before 34+0 and 37+0 weeks. Data analysis allowed measure of Actim® Partus test sensitivity (Se), specificity (Sp), positive and negative predictive values (PPV and NPV), diagnostic efficiency as well as positive and negative likelihood ratios. Actim® Partus test features (Se, Sp, PPV and NPV) were 53.3, 67.9, 23.5 and 88.7% respectively for deliveries occurring ≤ 34+0 weeks and 54.2, 75.4, 55.8, and 74.2%, respectively, for deliveries occurring ≤ 37+0 weeks. Diagnostic efficiency of the test was 65.7% (≤ 34+0 weeks) and 67.7% (≤ 37+0 weeks). Positive likelihood ratios were 1.6 (≤ 34+0 weeks) and 2.2 (≤ 37+0 weeks). Negative likelihood ratios were 0.7 (≤ 34+0 weeks) and 0.6 (≤ 37+0 weeks). Results of our study show that phIGFBP-1 diagnostic test is not accurate enough in predicting preterm birth before 34+0 or 37+0 weeks, and therefore, there is little clinical interest in its everyday use.


Assuntos
Colo do Útero/metabolismo , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Trabalho de Parto Prematuro/diagnóstico , Nascimento Prematuro/diagnóstico , Vagina/metabolismo , Adulto , Feminino , Idade Gestacional , Humanos , Serviços de Saúde Materna , Trabalho de Parto Prematuro/metabolismo , Fosforilação , Valor Preditivo dos Testes , Gravidez , Nascimento Prematuro/metabolismo , Estudos Retrospectivos , Sensibilidade e Especificidade
9.
Eur J Neurosci ; 32(6): 905-20, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20726889

RESUMO

Alzheimer's disease (AD) affects cognitive modalities that are known to be regulated by adult neurogenesis, such as hippocampal- and olfactory-dependent learning and memory. However, the relationship between AD-associated pathologies and alterations in adult neurogenesis has remained contentious. In the present study, we performed a detailed investigation of adult neurogenesis in the triple transgenic (3xTg) mouse model of AD, a unique model that generates both amyloid plaques and neurofibrillary tangles, the hallmark pathologies of AD. In both neurogenic niches of the brain, the hippocampal dentate gyrus and forebrain subventricular zone, we found that 3xTg mice had decreased numbers of (i) proliferating cells, (ii) early lineage neural progenitors, and (iii) neuroblasts at middle age (11months old) and old age (18months old). These decreases correlated with major reductions in the addition of new neurons to the respective target areas, the dentate granule cell layer and olfactory bulb. Within the subventricular zone niche, cytological alterations were observed that included a selective loss of subependymal cells and the development of large lipid droplets within the ependyma of 3xTg mice, indicative of metabolic changes. Temporally, there was a marked acceleration of age-related decreases in 3xTg mice, which affected multiple stages of neurogenesis and was clearly apparent prior to the development of amyloid plaques or neurofibrillary tangles. Our findings indicate that AD-associated mutations suppress neurogenesis early during disease development. This suggests that deficits in adult neurogenesis may mediate premature cognitive decline in AD.


Assuntos
Doença de Alzheimer/patologia , Modelos Animais de Doenças , Emaranhados Neurofibrilares/patologia , Neurogênese/genética , Placa Amiloide/patologia , Fatores Etários , Doença de Alzheimer/genética , Animais , Proliferação de Células , Feminino , Técnicas de Introdução de Genes , Humanos , Camundongos , Camundongos Transgênicos , Emaranhados Neurofibrilares/genética , Placa Amiloide/genética
10.
Theranostics ; 10(14): 6337-6360, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32483456

RESUMO

Rationale: Monoacylglycerol lipase (Mgll), a hydrolase that breaks down the endocannabinoid 2-arachidonoyl glycerol (2-AG) to produce arachidonic acid (ARA), is a potential target for neurodegenerative diseases, such as Alzheimer's disease (AD). Increasing evidence shows that impairment of adult neurogenesis by perturbed lipid metabolism predisposes patients to AD. However, it remains unknown what causes aberrant expression of Mgll in AD and how Mgll-regulated lipid metabolism impacts adult neurogenesis, thus predisposing to AD during aging. Here, we identify Mgll as an aging-induced factor that impairs adult neurogenesis and spatial memory in AD, and show that metformin, an FDA-approved anti-diabetic drug, can reduce the expression of Mgll to reverse impaired adult neurogenesis, prevent spatial memory decline and reduce ß-amyloid accumulation. Methods: Mgll expression was assessed in both human AD patient post-mortem hippocampal tissues and 3xTg-AD mouse model. In addition, we used both the 3xTg-AD animal model and the CbpS436A genetic knock-in mouse model to identify that elevated Mgll expression is caused by the attenuation of the aPKC-CBP pathway, involving atypical protein kinase C (aPKC)-stimulated Ser436 phosphorylation of histone acetyltransferase CBP through biochemical methods. Furthermore, we performed in vivo adult neurogenesis assay with BrdU/EdU labelling and Morris water maze task in both animal models following pharmacological treatments to show the key role of Mgll in metformin-corrected neurogenesis and spatial memory deficits of AD through reactivating the aPKC-CBP pathway. Finally, we performed in vitro adult neurosphere assays using both animal models to study the role of the aPKC-CBP mediated Mgll repression in determining adult neural stem/progenitor cell (NPC) fate. Results: Here, we demonstrate that aging-dependent induction of Mgll is observed in the 3xTg-AD model and human AD patient post-mortem hippocampal tissues. Importantly, we discover that elevated Mgll expression is caused by the attenuation of the aPKC-CBP pathway. The accumulation of Mgll in the 3xTg-AD mice reduces the genesis of newborn neurons and perturbs spatial memory. However, we find that metformin-stimulated aPKC-CBP pathway decreases Mgll expression to recover these deficits in 3xTg-AD. In addition, we reveal that elevated Mgll levels in cultured adult NPCs from both 3xTg-AD and CbpS436A animal models are responsible for their NPC neuronal differentiation deficits. Conclusion: Our findings set the stage for development of a clinical protocol where Mgll would serve as a biomarker in early stages of AD to identify potential metformin-responsive AD patients to restore their neurogenesis and spatial memory.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/enzimologia , Metformina/farmacologia , Monoacilglicerol Lipases/metabolismo , Neurogênese/efeitos dos fármacos , Memória Espacial/efeitos dos fármacos , Doença de Alzheimer/patologia , Animais , Biomarcadores/metabolismo , Proteína de Ligação a CREB/metabolismo , Modelos Animais de Doenças , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Humanos , Hipoglicemiantes/farmacologia , Masculino , Camundongos , Camundongos Transgênicos , Proteína Quinase C/metabolismo
11.
Life Sci Alliance ; 3(7)2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32482782

RESUMO

The ventricular epithelium of the adult forebrain is a heterogeneous cell population that is a source of both quiescent and activated neural stem cells (qNSCs and aNSCs, respectively). We genetically targeted a subset of ventricle-contacting, glial fibrillary acidic protein (GFAP)-expressing cells, to study their involvement in qNSC/aNSC-mediated adult neurogenesis. Ventricle-contacting GFAP+ cells were lineage-traced beginning in early adulthood using adult brain electroporation and produced small numbers of olfactory bulb neuroblasts until at least 21 mo of age. Notably, electroporated GFAP+ neurogenic precursors were distinct from both qNSCs and aNSCs: they did not give rise to neurosphere-forming aNSCs in vivo or after extended passaging in vitro and they were not recruited during niche regeneration. GFAP+ cells with these properties included a FoxJ1+GFAP+ subset, as they were also present in an inducible FoxJ1 transgenic lineage-tracing model. Transiently overexpressing Mash1 increased the neurogenic output of electroporated GFAP+ cells in vivo, identifying them as a potentially recruitable population. We propose that the qNSC/aNSC lineage of the adult forebrain coexists with a distinct, minimally expanding subset of GFAP+ neurogenic precursors.


Assuntos
Ventrículos Cerebrais/metabolismo , Epitélio/metabolismo , Marcação de Genes , Fatores de Crescimento Neural/genética , Células-Tronco Neurais/metabolismo , Prosencéfalo/metabolismo , Adulto , Células-Tronco Adultas/metabolismo , Animais , Biomarcadores , Diferenciação Celular/genética , Imunofluorescência , Expressão Gênica , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Fatores de Crescimento Neural/metabolismo , Células-Tronco Neurais/citologia , Neurogênese/genética , Neurônios/citologia , Neurônios/metabolismo , Bulbo Olfatório/citologia , Bulbo Olfatório/metabolismo , Nicho de Células-Tronco/genética
12.
Hippocampus ; 19(10): 913-27, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19405143

RESUMO

Voluntary wheel-running induces a rapid increase in proliferation and neurogenesis by neural precursors present in the adult rodent hippocampus. In contrast, the responses of hippocampal and other central nervous system neural precursors following longer periods of voluntary physical activity are unclear and are an issue of potential relevance to physical rehabilitation programs. We investigated the effects of a prolonged, 6-week voluntary wheel-running paradigm on neural precursors of the CD1 mouse hippocampus and forebrain. Examination of the hippocampus following 6 weeks of running revealed two to three times as many newly born neurons and 60% more proliferating cells when compared with standard-housed control mice. Among running mice, the number of newly born neurons correlated with the total running distance. To establish the effects of wheel-running on hippocampal precursors dividing during later stages of the prolonged running regime, BrdU was administered after 3 weeks of running and the BrdU-retaining cells were analyzed 18 days later. Quantifications revealed that the effects of wheel-running were maintained in late-stage proliferating cells, as running mice had two to three times as many BrdU-retaining cells within the hippocampal dentate gyrus, and these yielded greater proportions of both mature neurons and proliferative cells. The effects of prolonged wheel-running were also detected beyond the hippocampus. Unlike short-term wheel-running, prolonged wheel-running was associated with higher numbers of proliferating cells within the ventral forebrain subventricular region, a site of age-associated decreases in neural precursor proliferation and neurogenesis. Collectively, these findings indicate that (i) prolonged voluntary wheel-running maintains an increased level of hippocampal neurogenesis whose magnitude is linked to total running performance, and (ii) that it influences multiple neural precursor populations of the adult mouse brain.


Assuntos
Células-Tronco Adultas/fisiologia , Hipocampo/fisiologia , Neurogênese/fisiologia , Neurônios/fisiologia , Condicionamento Físico Animal/fisiologia , Prosencéfalo/fisiologia , Células-Tronco Adultas/citologia , Animais , Bromodesoxiuridina , Contagem de Células , Proliferação de Células , Hipocampo/citologia , Imuno-Histoquímica , Antígeno Ki-67/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos , Mitose/fisiologia , Neurônios/citologia , Prosencéfalo/citologia , Corrida/fisiologia , Nicho de Células-Tronco/fisiologia , Fatores de Tempo , Volição
13.
Neuron ; 34(6): 985-98, 2002 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-12086645

RESUMO

Neuronal activity and neurotrophins play a central role in the formation, maintenance, and plasticity of dendritic arbors. Here, we show that neuronal activity, mediated by electrical stimulation, KCl depolarization, or cholinergic receptor activation, promotes reversible dendrite formation in sympathetic neurons and that this effect is enhanced by NGF. Activity-dependent dendrite formation is accompanied by increased association of HMW MAP2 with microtubules and increased microtubule stability. Inhibition of either CaMKII or the MEK-ERK pathway, both of which phosphorylate MAP2, inhibits dendrite formation, but inhibition of both pathways simultaneously is required for dendrites to retract. These data indicate that neuronal activity signals via CamKII and the ERKs to regulate MAP2:microtubule interactions and hence reversible dendrite stability, and to provide a mechanism whereby activity and neurotrophins converge intracellularly to dynamically regulate dendritic morphology.


Assuntos
Dendritos/fisiologia , Transdução de Sinais/fisiologia , Fibras Adrenérgicas/efeitos dos fármacos , Fibras Adrenérgicas/enzimologia , Fibras Adrenérgicas/fisiologia , Animais , Animais Recém-Nascidos , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Células Cultivadas , Técnicas de Cultura , Dendritos/efeitos dos fármacos , Dendritos/enzimologia , Estimulação Elétrica/métodos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fator de Crescimento Neural/farmacologia , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Cloreto de Potássio/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Colinérgicos/metabolismo , Transdução de Sinais/efeitos dos fármacos
14.
J Mol Cell Cardiol ; 45(5): 694-702, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18718475

RESUMO

Nestin-expressing cells were identified in the normal rat heart characterized by a small cell body and numerous processes and following an ischemic insult migrated to the infarct region. The present study was undertaken to identify the phenotype, origin and biological role of nestin-expressing cells during reparative fibrosis. A neural stem cell phenotype was identified based on musashi-1 expression, growth as a neurosphere, and differentiation to a neuronal cell. Using the Wnt1-cre; Z/EG transgenic mouse model, which expresses EGFP in embryologically-derived neural crest cells, the reporter signal was detected in nestin-expressing cells residing in the heart. In infarcted human hearts, nestin-expressing cells were detected in the viable myocardium and the scar and morphologically analogous to the population identified in the rat heart. Following either an ischemic insult or the acute administration of 6-hydroxydopamine, sympathetic sprouting was dependent on the physical association of neurofilament-M immunoreactive fibres with nestin-positive processes emanating from neural stem cells. To specifically study the biological role of the subpopulation in the infarct region, neural stem cells were isolated from the scar, fluorescently labelled and transplanted in the heart of 3-day post-MI rats. Injected scar-derived neural stem cells migrated to the infarct region and were used as a substrate for de novo blood vessel formation. These data have demonstrated that the heart contains a resident population of neural stem cells derived from the neural crest and participate in reparative fibrosis. Their manipulation could provide an alternative approach to ameliorate the healing process following ischemic injury.


Assuntos
Coração/fisiologia , Neovascularização Fisiológica , Animais , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Miocárdio/metabolismo , Crista Neural/metabolismo , Proteínas de Neurofilamentos/metabolismo , Neurônios/metabolismo , Oxidopamina/farmacologia , Ratos , Ratos Sprague-Dawley , Células-Tronco/metabolismo
15.
Front Mol Neurosci ; 11: 126, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29706867

RESUMO

Environmental enrichment (EE) is a powerful stimulus of brain plasticity and is among the most accessible treatment options for brain disease. In rodents, EE is modeled using multi-factorial environments that include running, social interactions, and/or complex surroundings. Here, we show that running and running-independent EE differentially affect the hippocampal dentate gyrus (DG), a brain region critical for learning and memory. Outbred male CD1 mice housed individually with a voluntary running disk showed improved spatial memory in the radial arm maze compared to individually- or socially-housed mice with a locked disk. We therefore used RNA sequencing to perform an unbiased interrogation of DG gene expression in mice exposed to either a voluntary running disk (RUN), a locked disk (LD), or a locked disk plus social enrichment and tunnels [i.e., a running-independent complex environment (CE)]. RNA sequencing revealed that RUN and CE mice showed distinct, non-overlapping patterns of transcriptomic changes versus the LD control. Bio-informatics uncovered that the RUN and CE environments modulate separate transcriptional networks, biological processes, cellular compartments and molecular pathways, with RUN preferentially regulating synaptic and growth-related pathways and CE altering extracellular matrix-related functions. Within the RUN group, high-distance runners also showed selective stress pathway alterations that correlated with a drastic decline in overall transcriptional changes, suggesting that excess running causes a stress-induced suppression of running's genetic effects. Our findings reveal stimulus-dependent transcriptional signatures of EE on the DG, and provide a resource for generating unbiased, data-driven hypotheses for novel mediators of EE-induced cognitive changes.

17.
Front Neurosci ; 9: 407, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26576147

RESUMO

A single asymmetric division by an adult neural stem cell (NSC) ultimately generates dozens of differentiated progeny, a feat made possible by the proliferative expansion of transit-amplifying progenitor cells (TAPs). Although NSC activation and TAP expansion is determined by pro- and anti-proliferative signals found within the niche, remarkably little is known about how these cells integrate simultaneous conflicting signals. We investigated this question focusing on the subventricular zone (SVZ) niche of the adult murine forebrain. Using primary cultures of SVZ cells, we demonstrate that Epidermal Growth Factor (EGF) and Bone Morphogenetic Protein (BMP)-2 are particularly powerful pro- and anti-proliferative factors for SVZ-derived neural precursors. Dose-response experiments showed that when simultaneously exposed to both signals, BMP dominantly suppressed EGF-induced proliferation; moreover, this dominance extended to all parameters of neural precursor behavior tested, including inhibition of proliferation, modulation of cell cycle, promotion of differentiation, and increase of cell death. BMP's anti-proliferative effect did not involve inhibition of mTORC1 or ERK signaling, key mediators of EGF-induced proliferation, and had distinct stage-specific consequences, promoting TAP differentiation but NSC quiescence. In line with these in vitro data, in vivo experiments showed that exogenous BMP limits EGF-induced proliferation of TAPs while inhibition of BMP-SMAD signaling promotes activation of quiescent NSCs. These findings clarify the stage-specific effects of BMPs on SVZ neural precursors, and support a hierarchical model in which the anti-proliferative effects of BMP dominate over EGF proliferation signaling to constitutively drive TAP differentiation and NSC quiescence.

18.
Cell Stem Cell ; 17(4): 397-411, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26321199

RESUMO

Lipid metabolism is fundamental for brain development and function, but its roles in normal and pathological neural stem cell (NSC) regulation remain largely unexplored. Here, we uncover a fatty acid-mediated mechanism suppressing endogenous NSC activity in Alzheimer's disease (AD). We found that postmortem AD brains and triple-transgenic Alzheimer's disease (3xTg-AD) mice accumulate neutral lipids within ependymal cells, the main support cell of the forebrain NSC niche. Mass spectrometry and microarray analyses identified these lipids as oleic acid-enriched triglycerides that originate from niche-derived rather than peripheral lipid metabolism defects. In wild-type mice, locally increasing oleic acid was sufficient to recapitulate the AD-associated ependymal triglyceride phenotype and inhibit NSC proliferation. Moreover, inhibiting the rate-limiting enzyme of oleic acid synthesis rescued proliferative defects in both adult neurogenic niches of 3xTg-AD mice. These studies support a pathogenic mechanism whereby AD-induced perturbation of niche fatty acid metabolism suppresses the homeostatic and regenerative functions of NSCs.


Assuntos
Metabolismo dos Lipídeos , Células-Tronco Neurais , Prosencéfalo/metabolismo , Células-Tronco Adultas/metabolismo , Células-Tronco Adultas/patologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Autopsia , Proliferação de Células , Modelos Animais de Doenças , Espectrometria de Massas , Camundongos , Análise em Microsséries , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/patologia , Ácido Oleico/biossíntese , Regeneração , Nicho de Células-Tronco
19.
PLoS One ; 9(1): e86237, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24465980

RESUMO

Environmental enrichment (EE) exerts powerful effects on brain physiology, and is widely used as an experimental and therapeutic tool. Typical EE paradigms are multifactorial, incorporating elements of physical exercise, environmental complexity, social interactions and stress, however the specific contributions of these variables have not been separable using conventional housing paradigms. Here, we evaluated the impacts of these individual variables on adult hippocampal neurogenesis by using a novel "Alternating EE" paradigm. For 4 weeks, adult male CD1 mice were alternated daily between two enriched environments; by comparing groups that differed in one of their two environments, the individual and combinatorial effects of EE variables could be resolved. The Alternating EE paradigm revealed that (1) voluntary running for 3 days/week was sufficient to increase both mitotic and post-mitotic stages of hippocampal neurogenesis, confirming the central importance of exercise; (2) a complex environment (comprised of both social interactions and rotated inanimate objects) had no effect on neurogenesis itself, but enhanced depolarization-induced c-Fos expression (attributable to social interactions) and buffered stress-induced plasma corticosterone levels (attributable to inanimate objects); and (3) neither social isolation, group housing, nor chronically increased levels of plasma corticosterone had a prolonged impact on neurogenesis. Mouse strain, handling and type of running apparatus were tested and excluded as potential confounding factors. These findings provide valuable insights into the relative effects of key EE variables on adult neurogenesis, and this "Alternating EE" paradigm represents a useful tool for exploring the contributions of individual EE variables to mechanisms of neural plasticity.


Assuntos
Giro Denteado/fisiopatologia , Neurogênese , Corrida , Estresse Psicológico/fisiopatologia , Animais , Comportamento Animal , Corticosterona/sangue , Giro Denteado/metabolismo , Giro Denteado/patologia , Meio Ambiente , Expressão Gênica , Abrigo para Animais , Masculino , Potenciais da Membrana , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Comportamento Social , Estresse Psicológico/metabolismo , Estresse Psicológico/patologia
20.
Exp Neurol ; 201(1): 32-48, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16678161

RESUMO

Multipotent precursors similar to stem cells of the embryonic neural crest (NC) have been identified in several postnatal tissues, and are potentially useful for research and therapeutic purposes. However, their neurogenic potential, including their ability to produce electrophysiologically active neurons, is largely unexplored. We investigated this issue with regard to skin-derived precursors (SKPs), multipotent NC-related precursors isolated from the dermis of skin. SKP cultures follow an appropriate pattern and time-course of neuronal differentiation, with proliferating nestin-expressing SKPs generating post-mitotic neuronal cells that co-express pan-neuronal and peripheral autonomic lineage markers. These SKP-derived neuron-like cells survive and maintain their peripheral phenotype for at least 5 weeks when transplanted into the CNS environment of normal or kainate-injured hippocampal slices. Undifferentiated SKPs retain key neural precursor properties after multi-passage expansion, including growth factor dependence, nestin expression, neurogenic potential, and responsiveness to embryonic neural crest fate determinants. Despite undergoing an apparently appropriate neurogenic process, however, SKP-derived neuron-like cells possess an immature electrophysiological profile. These findings indicate that SKPs retain latent neurogenic properties after residing in a non-neural tissue, but that additional measures will be necessary to promote their differentiation into electrophysiologically active neurons.


Assuntos
Células-Tronco Multipotentes/citologia , Neurônios/citologia , Pele/citologia , Animais , Western Blotting , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Fator de Crescimento Epidérmico/farmacologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hipocampo/citologia , Hipocampo/metabolismo , Proteínas de Filamentos Intermediários/metabolismo , Glicoproteínas de Membrana/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Transgênicos , Células-Tronco Multipotentes/metabolismo , Células-Tronco Multipotentes/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Moléculas de Adesão de Célula Nervosa/metabolismo , Neurônios/metabolismo , Neurônios/fisiologia , Técnicas de Patch-Clamp , Periferinas , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Pele/efeitos dos fármacos , Pele/metabolismo , Fatores de Tempo , Técnicas de Cultura de Tecidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA