Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stem Cell Res ; 67: 103012, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36610307

RESUMO

C-terminal Binding Protein 1 (CTBP1) is a ubiquitously expressed transcriptional co-repressor and membrane trafficking regulator. A recurrent de novo c.991C>T mutation in CTBP1 leads to expression of p.R331W CTBP1 and causes hypotonia, ataxia, developmental delay, and tooth enamel defects syndrome (HADDTS), a rare early onset neurodevelopmental disorder. We generated hESCs lines with heterozygote and homozygote c.991C>T in CTBP1 using CRISPR/Cas9 genome editing and validated them for genetic integrity, off-target mutations, and pluripotency. They will be useful for investigation of HADDTS pathophysiology and for screening for potential therapeutics.


Assuntos
Células-Tronco Embrionárias Humanas , Humanos , Ataxia/genética , Sistemas CRISPR-Cas , Heterozigoto , Homozigoto , Hipotonia Muscular/genética , Mutação , Fatores de Transcrição/genética
2.
Mol Psychiatry ; 28(1): 497-514, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-35318461

RESUMO

The transcription factor FOXG1 serves pleiotropic functions in brain development ranging from the regulation of precursor proliferation to the control of cortical circuit formation. Loss-of-function mutations and duplications of FOXG1 are associated with neurodevelopmental disorders in humans illustrating the importance of FOXG1 dosage for brain development. Aberrant FOXG1 dosage has been found to disrupt the balanced activity of glutamatergic and GABAergic neurons, but the underlying mechanisms are not fully understood. We report that FOXG1 is expressed in the main adult neurogenic niches in mice, i.e. the hippocampal dentate gyrus and the subependymal zone/olfactory bulb system, where neurogenesis of glutamatergic and GABAergic neurons persists into adulthood. These niches displayed differential vulnerability to increased FOXG1 dosage: high FOXG1 levels severely compromised survival and glutamatergic dentate granule neuron fate acquisition in the hippocampal neurogenic niche, but left neurogenesis of GABAergic neurons in the subependymal zone/olfactory bulb system unaffected. Comparative transcriptomic analyses revealed a significantly higher expression of the apoptosis-linked nuclear receptor Nr4a1 in FOXG1-overexpressing hippocampal neural precursors. Strikingly, pharmacological interference with NR4A1 function rescued FOXG1-dependent death of hippocampal progenitors. Our results reveal differential vulnerability of neuronal subtypes to increased FOXG1 dosage and suggest that activity of a FOXG1/NR4A1 axis contributes to such subtype-specific response.


Assuntos
Proteínas do Tecido Nervoso , Transtornos do Neurodesenvolvimento , Animais , Camundongos , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Hipocampo/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Transtornos do Neurodesenvolvimento/metabolismo , Neurogênese/genética , Neurônios/metabolismo , Humanos
3.
BMC Neurosci ; 21(1): 50, 2020 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-33228529

RESUMO

BACKGROUND: Transcription factor 4 (TCF4) has been linked to human neurodevelopmental disorders such as intellectual disability, Pitt-Hopkins Syndrome (PTHS), autism, and schizophrenia. Recent work demonstrated that TCF4 participates in the control of a wide range of neurodevelopmental processes in mammalian nervous system development including neural precursor proliferation, timing of differentiation, migration, dendritogenesis and synapse formation. TCF4 is highly expressed in the adult hippocampal dentate gyrus - one of the few brain regions where neural stem / progenitor cells generate new functional neurons throughout life. RESULTS: We here investigated whether TCF4 haploinsufficiency, which in humans causes non-syndromic forms of intellectual disability and PTHS, affects adult hippocampal neurogenesis, a process that is essential for hippocampal plasticity in rodents and potentially in humans. Young adult Tcf4 heterozygote knockout mice showed a major reduction in the level of adult hippocampal neurogenesis, which was at least in part caused by lower stem/progenitor cell numbers and impaired maturation and survival of adult-generated neurons. Interestingly, housing in an enriched environment was sufficient to enhance maturation and survival of new neurons and to substantially augment neurogenesis levels in Tcf4 heterozygote knockout mice. CONCLUSION: The present findings indicate that haploinsufficiency for the intellectual disability- and PTHS-linked transcription factor TCF4 not only affects embryonic neurodevelopment but impedes neurogenesis in the hippocampus of adult mice. These findings suggest that TCF4 haploinsufficiency may have a negative impact on hippocampal function throughout adulthood by impeding hippocampal neurogenesis.


Assuntos
Meio Ambiente , Haploinsuficiência/genética , Fator de Transcrição 4/deficiência , Fator de Transcrição 4/genética , Animais , Diferenciação Celular , Sobrevivência Celular , Fácies , Hipocampo/patologia , Hiperventilação , Deficiência Intelectual/genética , Camundongos , Camundongos Knockout , Neurogênese/genética , Neurônios/patologia
4.
Nucleic Acids Res ; 48(9): 4839-4857, 2020 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-32266943

RESUMO

Development of oligodendrocytes and myelin formation in the vertebrate central nervous system is under control of several basic helix-loop-helix transcription factors such as Olig2, Ascl1, Hes5 and the Id proteins. The class I basic helix-loop-helix proteins Tcf3, Tcf4 and Tcf12 represent potential heterodimerization partners and functional modulators for all, but have not been investigated in oligodendrocytes so far. Using mouse mutants, organotypic slice and primary cell cultures we here show that Tcf4 is required in a cell-autonomous manner for proper terminal differentiation and myelination in vivo and ex vivo. Partial compensation is provided by the paralogous Tcf3, but not Tcf12. On the mechanistic level Tcf4 was identified as the preferred heterodimerization partner of the central regulator of oligodendrocyte development Olig2. Both genetic studies in the mouse as well as functional studies on enhancer regions of myelin genes confirmed the relevance of this physical interaction for oligodendrocyte differentiation. Considering that alterations in TCF4 are associated with syndromic and non-syndromic forms of intellectual disability, schizophrenia and autism in humans, our findings point to the possibility of an oligodendroglial contribution to these disorders.


Assuntos
Fator de Transcrição 2 de Oligodendrócitos/genética , Oligodendroglia/citologia , Fator de Transcrição 4/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Linhagem Celular Tumoral , Células Cultivadas , Dimerização , Feminino , Deleção de Genes , Células HEK293 , Humanos , Masculino , Camundongos , Bainha de Mielina/fisiologia , Oligodendroglia/metabolismo , Ratos Wistar
5.
Cereb Cortex ; 30(6): 3731-3743, 2020 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-32080705

RESUMO

Neuronal activity initiates transcriptional programs that shape long-term changes in plasticity. Although neuron subtypes differ in their plasticity response, most activity-dependent transcription factors (TFs) are broadly expressed across neuron subtypes and brain regions. Thus, how region- and neuronal subtype-specific plasticity are established on the transcriptional level remains poorly understood. We report that in young adult (i.e., 6-8 weeks old) mice, the developmental TF SOX11 is induced in neurons within 6 h either by electroconvulsive stimulation or by exploration of a novel environment. Strikingly, SOX11 induction was restricted to the dentate gyrus (DG) of the hippocampus. In the novel environment paradigm, SOX11 was observed in a subset of c-FOS expressing neurons (ca. 15%); whereas around 75% of SOX11+ DG granule neurons were c-FOS+, indicating that SOX11 was induced in an activity-dependent fashion in a subset of neurons. Environmental enrichment or virus-mediated overexpression of SOX11 enhanced the excitability of DG granule cells and downregulated the expression of different potassium channel subunits, whereas conditional Sox11/4 knock-out mice presented the opposite phenotype. We propose that Sox11 is regulated in an activity-dependent fashion, which is specific to the DG, and speculate that activity-dependent Sox11 expression may participate in the modulation of DG neuron plasticity.


Assuntos
Giro Denteado/metabolismo , Comportamento Exploratório/fisiologia , Regulação da Expressão Gênica , Plasticidade Neuronal/genética , Neurônios/metabolismo , Fatores de Transcrição SOXC/genética , Animais , Eletrochoque , Camundongos , Camundongos Knockout , Técnicas de Patch-Clamp , Proteínas Proto-Oncogênicas c-fos/metabolismo , Fatores de Transcrição SOXC/metabolismo
6.
Front Mol Neurosci ; 12: 40, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30853890

RESUMO

Mitochondria are key organelles in regulating the metabolic state of a cell. In the brain, mitochondrial oxidative metabolism is the prevailing mechanism for neurons to generate ATP. While it is firmly established that neuronal function is highly dependent on mitochondrial metabolism, it is less well-understood how astrocytes function rely on mitochondria. In this study, we investigate if astrocytes require a functional mitochondrial electron transport chain (ETC) and oxidative phosphorylation (oxPhos) under physiological and injury conditions. By immunohistochemistry we show that astrocytes expressed components of the ETC and oxPhos complexes in vivo. Genetic inhibition of mitochondrial transcription by conditional deletion of mitochondrial transcription factor A (Tfam) led to dysfunctional ETC and oxPhos activity, as indicated by aberrant mitochondrial swelling in astrocytes. Mitochondrial dysfunction did not impair survival of astrocytes, but caused a reactive gliosis in the cortex under physiological conditions. Photochemically initiated thrombosis induced ischemic stroke led to formation of hyperfused mitochondrial networks in reactive astrocytes of the perilesional area. Importantly, mitochondrial dysfunction significantly reduced the generation of new astrocytes and increased neuronal cell death in the perilesional area. These results indicate that astrocytes require a functional ETC and oxPhos machinery for proliferation and neuroprotection under injury conditions.

7.
Neuron ; 99(6): 1188-1203.e6, 2018 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-30197237

RESUMO

Autophagy is a conserved catabolic pathway with emerging functions in mammalian neurodevelopment and human neurodevelopmental diseases. The mechanisms controlling autophagy in neuronal development are not fully understood. Here, we found that conditional deletion of the Forkhead Box O transcription factors FoxO1, FoxO3, and FoxO4 strongly impaired autophagic flux in developing neurons of the adult mouse hippocampus. Moreover, FoxO deficiency led to altered dendritic morphology, increased spine density, and aberrant spine positioning in adult-generated neurons. Strikingly, pharmacological induction of autophagy was sufficient to correct abnormal dendrite and spine development of FoxO-deficient neurons. Collectively, these findings reveal a novel link between FoxO transcription factors, autophagic flux, and maturation of developing neurons.


Assuntos
Autofagia/fisiologia , Fatores de Transcrição Forkhead/metabolismo , Morfogênese/fisiologia , Neurogênese/fisiologia , Animais , Separação Celular/métodos , Células Cultivadas , Camundongos Transgênicos , Neurônios/metabolismo
8.
Sci Rep ; 8(1): 9241, 2018 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-29915186

RESUMO

Neuronal production from neural stem cells persists during adulthood in the subgranular zone of the hippocampal dentate gyrus. Extracellular signals provided by the hippocampal microenvironment regulate the neuronal fate commitment of the stem cell progeny. To date, the identity of those signals and their crosstalk has been only partially resolved. Here we show that adult rat hippocampal neural stem and progenitor cells (AH-NSPCs) express receptors for bone morphogenetic proteins (BMPs) and that the BMP/P-Smad pathway is active in AH-NSPCs undergoing differentiation towards the neuronal lineage. In vitro, exposure to the BMP2 and BMP4 ligands is sufficient to increase neurogenesis from AH-NSPCs in a WNT dependent manner while decreasing oligodendrogenesis. Moreover, BMP2/4 and WNT3A, a key regulator of adult hippocampal neurogenesis, cooperate to further enhance neuronal production. Our data point to a mechanistic convergence of the BMP and WNT pathways at the level of the T-cell factor/lymphoid enhancer factor gene Lef1. Altogether, we provide evidence that BMP signalling is an important regulator for the neuronal fate specification of AH-NSPCs cultures and we show that it significantly cooperates with the previously described master regulator of adult hippocampal neurogenesis, the WNT signalling pathway.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Proteína Morfogenética Óssea 4/metabolismo , Hipocampo/metabolismo , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt/fisiologia , Animais , Diferenciação Celular/fisiologia , Giro Denteado/metabolismo , Camundongos , Neurogênese/fisiologia , Ratos , Fatores de Transcrição TCF/metabolismo
9.
J Neurochem ; 146(3): 251-268, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29749639

RESUMO

The high-mobility-group domain containing SoxC transcription factors Sox4 and Sox11 are expressed and required in the vertebrate central nervous system in neuronal precursors and neuroblasts. To identify genes that are widely regulated by SoxC proteins during vertebrate neurogenesis we generated expression profiles from developing mouse brain and chicken neural tube with reduced SoxC expression and found the transcription factor prospero homeobox protein 1 (Prox1) strongly down-regulated under both conditions. This led us to hypothesize that Prox1 expression depends on SoxC proteins in the developing central nervous system of mouse and chicken. By combining luciferase reporter assays and over-expression in the chicken neural tube with in vivo and in vitro binding studies, we identify the Prox1 gene promoter and two upstream enhancers at -44 kb and -40 kb relative to the transcription start as regulatory regions that are bound and activated by SoxC proteins. This argues that Prox1 is a direct target gene of SoxC proteins during neurogenesis. Electroporations in the chicken neural tube furthermore show that Prox1 activates a subset of SoxC target genes, whereas it has no effects on others. We propose that the transcriptional control of Prox1 by SoxC proteins may ensure coupling of two types of transcription factors that are both required during early neurogenesis, but have at least in part distinct functions. Open Data: Materials are available on https://cos.io/our-services/open-science-badges/ https://osf.io/93n6m/.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Homeodomínio/metabolismo , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Prosencéfalo/citologia , Fatores de Transcrição SOXC/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Embrião de Galinha , Imunoprecipitação da Cromatina , Biologia Computacional , Ensaio de Desvio de Mobilidade Eletroforética , Eletroporação , Embrião de Mamíferos , Ontologia Genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Tubo Neural/citologia , Tubo Neural/metabolismo , Fatores do Domínio POU/genética , Fatores do Domínio POU/metabolismo , Prosencéfalo/embriologia , Prosencéfalo/crescimento & desenvolvimento , Prosencéfalo/metabolismo , Fatores de Transcrição SOXC/genética , Tubulina (Proteína)/metabolismo , Proteínas Supressoras de Tumor/genética
10.
Mol Autism ; 9: 20, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29588831

RESUMO

Background: Haploinsufficiency of the class I bHLH transcription factor TCF4 causes Pitt-Hopkins syndrome (PTHS), a severe neurodevelopmental disorder, while common variants in the TCF4 gene have been identified as susceptibility factors for schizophrenia. It remains largely unknown, which brain regions are dependent on TCF4 for their development and function. Methods: We systematically analyzed the expression pattern of TCF4 in the developing and adult mouse brain. We used immunofluorescent staining to identify candidate regions whose development and function depend on TCF4. In addition, we determined TCF4 expression in the developing rhesus monkey brain and in the developing and adult human brain through analysis of transcriptomic datasets and compared the expression pattern between species. Finally, we morphometrically and histologically analyzed selected brain structures in Tcf4-haploinsufficient mice and compared our morphometric findings to neuroanatomical findings in PTHS patients. Results: TCF4 is broadly expressed in cortical and subcortical structures in the developing and adult mouse brain. The TCF4 expression pattern was highly similar between humans, rhesus monkeys, and mice. Moreover, Tcf4 haploinsufficiency in mice replicated structural brain anomalies observed in PTHS patients. Conclusion: Our data suggests that TCF4 is involved in the development and function of multiple brain regions and indicates that its regulation is evolutionary conserved. Moreover, our data validate Tcf4-haploinsufficient mice as a model to study the neurodevelopmental basis of PTHS.


Assuntos
Córtex Cerebral/metabolismo , Haploinsuficiência , Hipocampo/metabolismo , Hiperventilação/genética , Deficiência Intelectual/genética , Esquizofrenia/genética , Fator de Transcrição 4/genética , Animais , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/crescimento & desenvolvimento , Criança , Fácies , Feminino , Hipocampo/citologia , Hipocampo/crescimento & desenvolvimento , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Plasticidade Neuronal , Neurônios/metabolismo , Neurônios/fisiologia , Fator de Transcrição 4/metabolismo
11.
Cell Tissue Res ; 371(1): 91-103, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29079881

RESUMO

During development, generation of neurons is coordinated by the sequential activation of gene expression programs by stage- and subtype-specific transcription factor networks. The SoxC group transcription factors, Sox4 and Sox11, have recently emerged as critical components of this network. Initially identified as survival and differentiation factors for neural precursors, SoxC factors have now been linked to a broader array of developmental processes including neuronal subtype specification, migration, dendritogenesis and establishment of neuronal projections, and are now being employed in experimental strategies for neuronal replacement and axonal regeneration in the diseased central nervous system. This review summarizes the current knowledge regarding SoxC factor function in CNS development and disease and their promise for regeneration.


Assuntos
Encéfalo/embriologia , Neurogênese/fisiologia , Neurônios/fisiologia , Fatores de Transcrição SOXC/metabolismo , Animais , Reprogramação Celular , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Modelos Animais , Regeneração
13.
Neuron ; 93(3): 560-573.e6, 2017 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-28111078

RESUMO

Precise regulation of cellular metabolism is hypothesized to constitute a vital component of the developmental sequence underlying the life-long generation of hippocampal neurons from quiescent neural stem cells (NSCs). The identity of stage-specific metabolic programs and their impact on adult neurogenesis are largely unknown. We show that the adult hippocampal neurogenic lineage is critically dependent on the mitochondrial electron transport chain and oxidative phosphorylation machinery at the stage of the fast proliferating intermediate progenitor cell. Perturbation of mitochondrial complex function by ablation of the mitochondrial transcription factor A (Tfam) reproduces multiple hallmarks of aging in hippocampal neurogenesis, whereas pharmacological enhancement of mitochondrial function ameliorates age-associated neurogenesis defects. Together with the finding of age-associated alterations in mitochondrial function and morphology in NSCs, these data link mitochondrial complex function to efficient lineage progression of adult NSCs and identify mitochondrial function as a potential target to ameliorate neurogenesis-defects in the aging hippocampus.


Assuntos
Células-Tronco Adultas/metabolismo , Envelhecimento/metabolismo , Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Mitocôndrias/metabolismo , Neurogênese , Neurônios/metabolismo , Células-Tronco Adultas/citologia , Animais , Linhagem da Célula , Proliferação de Células , Células Cultivadas , Proteínas de Ligação a DNA/genética , Proteínas de Grupo de Alta Mobilidade/genética , Hipocampo/citologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Células-Tronco Neurais , Neurônios/citologia , Fosforilação Oxidativa
15.
Nat Commun ; 6: 8466, 2015 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-26506265

RESUMO

As human life expectancy has improved rapidly in industrialized societies, age-related cognitive impairment presents an increasing challenge. Targeting histopathological processes that correlate with age-related cognitive declines, such as neuroinflammation, low levels of neurogenesis, disrupted blood-brain barrier and altered neuronal activity, might lead to structural and functional rejuvenation of the aged brain. Here we show that a 6-week treatment of young (4 months) and old (20 months) rats with montelukast, a marketed anti-asthmatic drug antagonizing leukotriene receptors, reduces neuroinflammation, elevates hippocampal neurogenesis and improves learning and memory in old animals. By using gene knockdown and knockout approaches, we demonstrate that the effect is mediated through inhibition of the GPR17 receptor. This work illustrates that inhibition of leukotriene receptor signalling might represent a safe and druggable target to restore cognitive functions in old individuals and paves the way for future clinical translation of leukotriene receptor inhibition for the treatment of dementias.


Assuntos
Envelhecimento/efeitos dos fármacos , Antiasmáticos/administração & dosagem , Encéfalo/efeitos dos fármacos , Adulto , Fatores Etários , Envelhecimento/fisiologia , Animais , Encéfalo/fisiologia , Cognição , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Humanos , Masculino , Aprendizagem em Labirinto , Neurônios/citologia , Neurônios/metabolismo , Ratos , Ratos Endogâmicos F344 , Receptores de Leucotrienos/genética , Receptores de Leucotrienos/metabolismo , Adulto Jovem
16.
BMC Neurosci ; 16: 60, 2015 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-26386671

RESUMO

BACKGROUND: Neurogenesis in the brain of adult mammals occurs throughout life in two locations: the subventricular zone of the lateral ventricle and the subgranular zone of the dentate gyrus in the hippocampus. RNA interference mechanisms have emerged as critical regulators of neuronal differentiation. However, to date, little is known about its function in adult neurogenesis. RESULTS: Here we show that the RNA interference machinery regulates Doublecortin levels and is associated with chromatin in differentiating adult neural progenitors. Deletion of Dicer causes abnormal higher levels of Doublecortin. The microRNA pathway plays an important role in Doublecortin regulation. In particular miRNA-128 overexpression can reduce Doublecortin levels in differentiating adult neural progenitors. CONCLUSIONS: We conclude that the RNA interference components play an important role, even through chromatin association, in regulating neuron-specific gene expression programs.


Assuntos
RNA Helicases DEAD-box/metabolismo , Expressão Gênica/fisiologia , Hipocampo/metabolismo , MicroRNAs/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese/fisiologia , Neuropeptídeos/metabolismo , Interferência de RNA/fisiologia , Ribonuclease III/metabolismo , Animais , Cromatina/metabolismo , RNA Helicases DEAD-box/genética , Proteínas do Domínio Duplacortina , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ribonuclease III/genética
17.
J Neurosci ; 34(19): 6624-33, 2014 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-24806687

RESUMO

Neural stem cells in the adult mammalian hippocampus continuously generate new functional neurons, which modify the hippocampal network and significantly contribute to cognitive processes and mood regulation. Here, we show that the development of new neurons from stem cells in adult mice is paralleled by extensive changes to mitochondrial mass, distribution, and shape. Moreover, exercise-a strong modifier of adult hippocampal neurogenesis-accelerates neuronal maturation and induces a profound increase in mitochondrial content and the presence of mitochondria in dendritic segments. Genetic inhibition of the activity of the mitochondrial fission factor dynamin-related protein 1 (Drp1) inhibits neurogenesis under basal and exercise conditions. Conversely, enhanced Drp1 activity furthers exercise-induced acceleration of neuronal maturation. Collectively, these results indicate that adult hippocampal neurogenesis requires adaptation of the mitochondrial compartment and suggest that mitochondria are targets for enhancing neurogenesis-dependent hippocampal plasticity.


Assuntos
Encéfalo/citologia , Encéfalo/crescimento & desenvolvimento , Mitocôndrias/fisiologia , Células-Tronco Neurais/fisiologia , Condicionamento Físico Animal/fisiologia , Animais , Contagem de Células , Diferenciação Celular/fisiologia , Dendritos/fisiologia , Dendritos/ultraestrutura , Espinhas Dendríticas/fisiologia , Espinhas Dendríticas/ultraestrutura , Dinaminas/biossíntese , Dinaminas/genética , Feminino , Hipocampo/citologia , Hipocampo/crescimento & desenvolvimento , Hipocampo/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Neurogênese/genética , Neurogênese/fisiologia , Técnicas Estereotáxicas
18.
Mamm Genome ; 24(9-10): 333-48, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24096375

RESUMO

ßB2-crystallin (gene symbol: Crybb2/CRYBB2) was first described as a structural protein of the ocular lens. This gene, however, is also expressed in several regions of the mammalian brain, although its function in this organ remains entirely unknown. To unravel some aspects of its function in the brain, we combined behavioral, neuroanatomical, and physiological analyses in a novel Crybb2 mouse mutant, O377. Behavioral tests with male O377 mutants revealed altered sensorimotor gating, suggesting modified neuronal functions. Since these mouse mutants also displayed reduced hippocampal size, we concentrated further investigations on the hippocampus. Free intracellular Ca(2+) levels were increased and apoptosis was enhanced in the hippocampus of O377 mutants. Moreover, the expression of the gene encoding calpain 3 (gene symbol Capn3) was elevated and the expression of genes coding for the NMDA receptor subunits was downregulated. Additionally, the number of parvalbumin-positive interneurons was decreased in the hippocampus but not in the cortex of the mutants. High-speed voltage-sensitive dye imaging demonstrated an increased translation of input-to-output neuronal activity in the dentate gyrus of this Crybb2 mutant. These results point to an important function of ßB2-crystallin in the hippocampal network. They indicate pleiotropic effects of mutations in the Crybb2 gene, which previously had been considered to be specific to the ocular lens. Moreover, our results are the first to demonstrate that ßB2-crystallin has a role in hippocampal function and behavioral phenotypes. This model can now be further explored by future experiments.


Assuntos
Giro Denteado/metabolismo , Filtro Sensorial , Cadeia B de beta-Cristalina/metabolismo , Animais , Apoptose , Cálcio/metabolismo , Giro Denteado/patologia , Giro Denteado/fisiopatologia , Comportamento Exploratório , Homeostase , Homozigoto , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Tamanho do Órgão , Fenótipo , Cadeia B de beta-Cristalina/genética
19.
PLoS One ; 8(5): e62693, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23667508

RESUMO

In mammals, adult neural stem cells give rise to new hippocampal dentate granule neurons and interneurons of the olfactory bulb throughout life. The microtubule associated protein Doublecortin (DCX) is expressed by migrating neuroblasts and immature neurons, and is widely used as a stage-specific marker of adult neurogenesis and as a marker to identify neurogenic activity in the adult brain per se. Mutations in the DCX gene have been causally linked to human lissencephalic syndromes. Moreover, embryonic loss of DCX function interferes with neuronal migration and dendritic patterning in a species- and region-specific manner. A putative function of DCX in adult neurogenesis has not been directly explored. Here we show that overexpression of DCX in newly generated dentate granule neurons of the adult mouse brain has no effect on morphological maturation or migration. We also show that micro (mi) RNA-mediated retroviral knockdown of DCX does not alter morphological maturation of adult born dentate granule cells or migration of new neurons in either adult neurogenic niche. Thus, the present data indicate that DCX is dispensable for the development of new neurons in adult mice.


Assuntos
Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Animais , Giro Denteado/citologia , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Feminino , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Ventrículos Laterais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/deficiência , Proteínas Associadas aos Microtúbulos/genética , Neurogênese , Neuropeptídeos/deficiência , Neuropeptídeos/genética , Retroviridae/genética
20.
Cell Stem Cell ; 11(4): 471-6, 2012 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-23040476

RESUMO

Reprogramming of somatic cells into neurons provides a new approach toward cell-based therapy of neurodegenerative diseases. A major challenge for the translation of neuronal reprogramming into therapy is whether the adult human brain contains cell populations amenable to direct somatic cell conversion. Here we show that cells from the adult human cerebral cortex expressing pericyte hallmarks can be reprogrammed into neuronal cells by retrovirus-mediated coexpression of the transcription factors Sox2 and Mash1. These induced neuronal cells acquire the ability of repetitive action potential firing and serve as synaptic targets for other neurons, indicating their capability of integrating into neural networks. Genetic fate-mapping in mice expressing an inducible Cre recombinase under the tissue-nonspecific alkaline phosphatase promoter corroborated the pericytic origin of the reprogrammed cells. Our results raise the possibility of functional conversion of endogenous cells in the adult human brain to induced neuronal fates.


Assuntos
Reprogramação Celular , Córtex Cerebral/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Neurais/citologia , Neurogênese , Neurônios/citologia , Pericitos/citologia , Potenciais de Ação , Adulto , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Células Cultivadas , Humanos , Camundongos , Rede Nervosa , Doenças Neurodegenerativas/terapia , Retroviridae , Fatores de Transcrição SOXB1/metabolismo , Transplante de Células-Tronco , Transmissão Sináptica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...