Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 216
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Annu Rev Cell Dev Biol ; 40(1): 381-406, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38985883

RESUMO

Neural stem cells (NSCs) persist in the adult mammalian brain and are able to give rise to new neurons and glia throughout life. The largest stem cell niche in the adult mouse brain is the ventricular-subventricular zone (V-SVZ) lining the lateral ventricles. Adult NSCs in the V-SVZ coexist in quiescent and actively proliferating states, and they exhibit a regionalized molecular identity. The importance of such spatial diversity is just emerging, as depending on their position within the niche, adult NSCs give rise to distinct subtypes of olfactory bulb interneurons and different types of glia. However, the functional relevance of stem cell heterogeneity in the V-SVZ is still poorly understood. Here, we put into perspective findings highlighting the importance of adult NSC diversity for brain plasticity, and how the body signals to brain stem cells in different physiological states to regulate their behavior.


Assuntos
Células-Tronco Adultas , Células-Tronco Neurais , Nicho de Células-Tronco , Animais , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Humanos , Ventrículos Laterais/citologia , Neurogênese , Bulbo Olfatório/citologia , Bulbo Olfatório/metabolismo , Camundongos , Encéfalo/citologia , Diferenciação Celular
2.
Annu Rev Cell Dev Biol ; 32: 127-141, 2016 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-27298094

RESUMO

The brain constantly changes to store memories and adapt to new conditions. One type of plasticity that has gained increasing interest during the last years is the generation of new cells. The generation of both new neurons and glial cells contributes to neural plasticity and to some neural repair. There are substantial differences between mammalian species with regard to the extent of and mechanisms behind cell exchange in neural plasticity. Both neurogenesis and gliogenesis have several specific features in humans, which may contribute to the unique plasticity of the human brain.


Assuntos
Regeneração Nervosa/fisiologia , Neurogênese , Neuroglia/citologia , Plasticidade Neuronal/fisiologia , Animais , Encéfalo/citologia , Humanos , Oligodendroglia/citologia
3.
EMBO J ; 2024 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-39300210

RESUMO

Astrocytes in the brain exhibit regional heterogeneity contributing to regional circuits involved in higher-order brain functions, yet the mechanisms controlling their distribution remain unclear. Here, we show that the precise allocation of astrocytes to specific brain regions during development is achieved through transcription factor 4 (Tcf4)-mediated fate restriction based on their embryonic origin. Loss of Tcf4 in ventral telencephalic neural progenitor cells alters the fate of oligodendrocyte precursor cells to transient intermediate astrocyte precursor cells, resulting in mislocalized astrocytes in the dorsal neocortex. These ectopic astrocytes engage with neocortical neurons and acquire features reminiscent of dorsal neocortical astrocytes. Furthermore, Tcf4 functions as a suppressor of astrocyte fate during the differentiation of oligodendrocyte precursor cells derived from the ventral telencephalon, thereby restricting the fate to the oligodendrocyte lineage in the dorsal neocortex. Together, our findings highlight a previously unappreciated role for Tcf4 in regulating astrocyte allocation, offering additional insights into the mechanisms underlying neurodevelopmental disorders linked to Tcf4 mutations.

4.
Proc Natl Acad Sci U S A ; 121(20): e2321711121, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38713624

RESUMO

During development, neural stem cells in the cerebral cortex, also known as radial glial cells (RGCs), generate excitatory neurons, followed by production of cortical macroglia and inhibitory neurons that migrate to the olfactory bulb (OB). Understanding the mechanisms for this lineage switch is fundamental for unraveling how proper numbers of diverse neuronal and glial cell types are controlled. We and others recently showed that Sonic Hedgehog (Shh) signaling promotes the cortical RGC lineage switch to generate cortical oligodendrocytes and OB interneurons. During this process, cortical RGCs generate intermediate progenitor cells that express critical gliogenesis genes Ascl1, Egfr, and Olig2. The increased Ascl1 expression and appearance of Egfr+ and Olig2+ cortical progenitors are concurrent with the switch from excitatory neurogenesis to gliogenesis and OB interneuron neurogenesis in the cortex. While Shh signaling promotes Olig2 expression in the developing spinal cord, the exact mechanism for this transcriptional regulation is not known. Furthermore, the transcriptional regulation of Olig2 and Egfr has not been explored. Here, we show that in cortical progenitor cells, multiple regulatory programs, including Pax6 and Gli3, prevent precocious expression of Olig2, a gene essential for production of cortical oligodendrocytes and astrocytes. We identify multiple enhancers that control Olig2 expression in cortical progenitors and show that the mechanisms for regulating Olig2 expression are conserved between the mouse and human. Our study reveals evolutionarily conserved regulatory logic controlling the lineage switch of cortical neural stem cells.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos , Córtex Cerebral , Receptores ErbB , Proteínas Hedgehog , Proteínas do Tecido Nervoso , Células-Tronco Neurais , Neurogênese , Fator de Transcrição 2 de Oligodendrócitos , Fator de Transcrição PAX6 , Animais , Neurogênese/fisiologia , Córtex Cerebral/metabolismo , Córtex Cerebral/citologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Receptores ErbB/metabolismo , Receptores ErbB/genética , Camundongos , Fator de Transcrição 2 de Oligodendrócitos/metabolismo , Fator de Transcrição 2 de Oligodendrócitos/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/genética , Fator de Transcrição PAX6/metabolismo , Fator de Transcrição PAX6/genética , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/citologia , Proteínas de Homeodomínio/metabolismo , Proteínas de Homeodomínio/genética , Proteína Gli3 com Dedos de Zinco/metabolismo , Proteína Gli3 com Dedos de Zinco/genética , Proteínas do Olho/metabolismo , Proteínas do Olho/genética , Proteínas Repressoras/metabolismo , Proteínas Repressoras/genética , Fatores de Transcrição Box Pareados/metabolismo , Fatores de Transcrição Box Pareados/genética , Neuroglia/metabolismo , Neuroglia/citologia , Regulação da Expressão Gênica no Desenvolvimento , Transdução de Sinais , Bulbo Olfatório/metabolismo , Bulbo Olfatório/citologia , Linhagem da Célula , Humanos
5.
Development ; 150(2)2023 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-36537580

RESUMO

Temporal identity factors regulate competence of neural progenitors to generate specific cell types in a time-dependent manner, but how they operate remains poorly defined. In the developing mouse retina, the Ikaros zinc-finger transcription factor Ikzf1 regulates production of early-born cell types, except cone photoreceptors. In this study we show that, during early stages of retinal development, another Ikaros family protein, Ikzf4, functions redundantly with Ikzf1 to regulate cone photoreceptor production. Using CUT&RUN and functional assays, we show that Ikzf4 binds and represses genes involved in late-born rod photoreceptor specification, hence favoring cone production. At late stages, when Ikzf1 is no longer expressed in progenitors, we show that Ikzf4 re-localizes to target genes involved in gliogenesis and is required for Müller glia production. We report that Ikzf4 regulates Notch signaling genes and is sufficient to activate the Hes1 promoter through two Ikzf GGAA-binding motifs, suggesting a mechanism by which Ikzf4 may influence gliogenesis. These results uncover a combinatorial role for Ikaros family members during nervous system development and provide mechanistic insights on how they temporally regulate cell fate output.


Assuntos
Fator de Transcrição Ikaros , Retina , Camundongos , Animais , Retina/metabolismo , Fator de Transcrição Ikaros/genética , Fator de Transcrição Ikaros/metabolismo , Células Fotorreceptoras Retinianas Cones/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Diferenciação Celular/genética
6.
Development ; 149(5)2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35253855

RESUMO

During mammalian brain development, how different astrocytes are specified from progenitor cells is not well understood. In particular, whether astrocyte progenitor cells (APCs) start as a relatively homogenous population or whether there is early heterogeneity remains unclear. Here, we have dissected subpopulations of embryonic mouse forebrain progenitors using single-cell transcriptome analyses. Our sequencing data revealed two molecularly distinct APC subgroups at the start of gliogenesis from both dorsal and ventral forebrains. The two APC subgroups were marked, respectively, by specific expression of Sparc and Sparcl1, which are known to function in mature astrocytes with opposing activities for regulating synapse formation. Expression analyses showed that SPARC and SPARCL1 mark APC subgroups that display distinct temporal and spatial patterns, correlating with major waves of astrogliogenesis during development. Our results uncover an early molecular divergence of APCs in the mammalian brain and provide a useful transcriptome resource for the study of glial cell specification.


Assuntos
Astrócitos/fisiologia , Mamíferos/fisiologia , Neurogênese/fisiologia , Neuroglia/fisiologia , Células-Tronco/fisiologia , Animais , Astrócitos/metabolismo , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Proliferação de Células/fisiologia , Mamíferos/metabolismo , Camundongos , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/fisiologia , Neuroglia/metabolismo , Osteonectina/metabolismo , Prosencéfalo/metabolismo , Prosencéfalo/fisiologia , Análise de Célula Única/métodos , Células-Tronco/metabolismo , Transcriptoma/fisiologia
7.
Int J Mol Sci ; 25(3)2024 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-38338702

RESUMO

In this review, we explore the intriguing realm of neurogenesis in the vestibular nuclei-a critical brainstem region governing balance and spatial orientation. We retrace almost 20 years of research into vestibular neurogenesis, from its discovery in the feline model in 2007 to the recent discovery of a vestibular neural stem cell niche. We explore the reasons why neurogenesis is important in the vestibular nuclei and the triggers for activating the vestibular neurogenic niche. We develop the symbiotic relationship between neurogenesis and gliogenesis to promote vestibular compensation. Finally, we examine the potential impact of reactive neurogenesis on vestibular compensation, highlighting its role in restoring balance through various mechanisms.


Assuntos
Núcleos Vestibulares , Vestíbulo do Labirinto , Gatos , Animais , Núcleos Vestibulares/patologia , Neurogênese , Células-Tronco , Tronco Encefálico
8.
J Neurosci ; 42(37): 7031-7046, 2022 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-35906071

RESUMO

Alpha-synuclein (αSyn) and tau are abundant multifunctional neuronal proteins, and their intracellular deposits have been linked to many neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease. Despite the disease relevance, their physiological roles remain elusive, as mice with knock-out of either of these genes do not exhibit overt phenotypes. To reveal functional cooperation, we generated αSyn-/-tau-/- double-knock-out mice and characterized the functional cross talk between these proteins during brain development. Intriguingly, deletion of αSyn and tau reduced Notch signaling and accelerated interkinetic nuclear migration of G2 phase at early embryonic stage. This significantly altered the balance between the proliferative and neurogenic divisions of progenitor cells, resulting in an overproduction of early born neurons and enhanced neurogenesis, by which the brain size was enlarged during the embryonic stage in both sexes. On the other hand, a reduction in the number of neural progenitor cells in the middle stage of corticogenesis diminished subsequent gliogenesis in the αSyn-/-tau-/- cortex. Additionally, the expansion and maturation of macroglial cells (astrocytes and oligodendrocytes) were suppressed in the αSyn-/-tau-/- postnatal brain, which in turn reduced the male αSyn-/-tau-/- brain size and cortical thickness to less than the control values. Our study identifies important functional cooperation of αSyn and tau during corticogenesis.SIGNIFICANCE STATEMENT Correct understanding of the physiological functions of αSyn and tau in CNS is critical to elucidate pathogenesis involved in the etiology of neurodegenerative diseases including Alzheimer's disease and Parkinson's disease. We show here that αSyn and tau are cooperatively involved in brain development via maintenance of progenitor cells. αSyn and tau double-knock-out mice exhibited an overproduction of early born neurons and accelerated neurogenesis at early corticogenesis. Furthermore, loss of αSyn and tau also perturbed gliogenesis at later embryonic stage, as well as the subsequent glial expansion and maturation at postnatal brain. Our findings provide new mechanistic insights and extend therapeutic opportunities for neurodegenerative diseases caused by aberrant αSyn and tau.


Assuntos
Doença de Alzheimer , Doenças Neurodegenerativas , Doença de Parkinson , Doença de Alzheimer/metabolismo , Animais , Feminino , Masculino , Camundongos , Camundongos Knockout , Doenças Neurodegenerativas/metabolismo , Doença de Parkinson/patologia , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
9.
EMBO Rep ; 22(11): e52728, 2021 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-34605607

RESUMO

During central nervous system development, neurogenesis and gliogenesis occur in an orderly manner to create precise neural circuitry. However, no systematic dataset of neural lineage development that covers both neurogenesis and gliogenesis for the human spinal cord is available. We here perform single-cell RNA sequencing of human spinal cord cells during embryonic and fetal stages that cover neuron generation as well as astrocytes and oligodendrocyte differentiation. We also map the timeline of sensory neurogenesis and gliogenesis in the spinal cord. We further identify a group of EGFR-expressing transitional glial cells with radial morphology at the onset of gliogenesis, which progressively acquires differentiated glial cell characteristics. These EGFR-expressing transitional glial cells exhibited a unique position-specific feature during spinal cord development. Cell crosstalk analysis using CellPhoneDB indicated that EGFR glial cells can persistently interact with other neural cells during development through Delta-Notch and EGFR signaling. Together, our results reveal stage-specific profiles and dynamics of neural cells during human spinal cord development.


Assuntos
Análise de Célula Única , Medula Espinal , Humanos , Neurogênese , Neuroglia , Neurônios
10.
Proc Natl Acad Sci U S A ; 117(36): 22214-22224, 2020 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-32848054

RESUMO

Increased neural stem cell (NSC) quiescence is a major determinant of age-related regenerative decline in the adult hippocampus. However, a coextensive model has been proposed in which division-coupled conversion of NSCs into differentiated astrocytes restrict the stem cell pool with age. Here we report that age-related loss of the posttranslational modification, O-linked ß-N-acetylglucosamine (O-GlcNAc), in NSCs promotes a glial fate switch. We detect an age-dependent decrease in NSC O-GlcNAc levels coincident with decreased neurogenesis and increased gliogenesis in the mature hippocampus. Mimicking an age-related loss of NSC O-GlcNAcylation in young mice reduces neurogenesis, increases astrocyte differentiation, and impairs associated cognitive function. Using RNA-sequencing of primary NSCs following decreased O-GlcNAcylation, we detected changes in the STAT3 signaling pathway indicative of glial differentiation. Moreover, using O-GlcNAc-specific mass spectrometry analysis of the aging hippocampus, together with an in vitro site-directed mutagenesis approach, we identify loss of STAT3 O-GlcNAc at Threonine 717 as a driver of astrocyte differentiation. Our data identify the posttranslational modification, O-GlcNAc, as a key molecular regulator of regenerative decline underlying an age-related NSC fate switch.


Assuntos
Envelhecimento/fisiologia , Diferenciação Celular/fisiologia , Glucosamina/análogos & derivados , Células-Tronco Neurais/fisiologia , Neuroglia/fisiologia , Fator de Transcrição STAT3/metabolismo , Animais , Proliferação de Células , Biologia Computacional , Regulação da Expressão Gênica , Glucosamina/metabolismo , Hipocampo/citologia , Camundongos , Neurogênese , Fator de Transcrição STAT3/genética , Análise de Sequência de RNA
11.
Differentiation ; 128: 13-25, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36198237

RESUMO

SOX10 gene and SOX10 protein are responsible for the gliogenesis of neuroglia from the neural crest cells. Expression of SOX10 gene encodes SOX10 protein which binds with DNA at its minor groove via its HMG domain upon activation. SOX10 protein undergoes bending and changes its conformation after binding with DNA. Via its transactivation domain and HMG domain, it further activates several other transcription factors, these cause gliogenesis of the neural crest cells into neuroglia. In literature, it is stated that the SOX10 gene helps in the formation of schwann cells, oligodendrocytes, and enteric ganglia from neural crest cells. Altered expression of the SOX10 gene results in agliogenesis, dysmyelination, and demyelination in the nervous system as well as intestinal aganglionosis. This review highlighted that there is a role of the SOX10 gene and SOX10 protein in enteric gliogenesis from the neural crest cells.


Assuntos
Sistema Nervoso Entérico , Doença de Hirschsprung , Humanos , Proteínas de Grupo de Alta Mobilidade/genética , Proteínas de Grupo de Alta Mobilidade/metabolismo , Doença de Hirschsprung/genética , Fatores de Transcrição SOXE/genética , Crista Neural , Sistema Nervoso Entérico/metabolismo
12.
Int J Mol Sci ; 24(16)2023 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-37629148

RESUMO

Numerous studies have focused on the pathophysiological role of amyloid precursor protein (APP) because the proteolytic processing of APP to ß-amyloid (Aß) peptide is a central event in Alzheimer's disease (AD). However, many authors consider that alterations in the physiological functions of APP are likely to play a key role in AD. Previous studies in our laboratory revealed that APP plays an important role in the differentiation of human neural stem cells (hNSCs), favoring glial differentiation (gliogenesis) and preventing their differentiation toward a neuronal phenotype (neurogenesis). In the present study, we have evaluated the effects of APP overexpression in hNSCs at a global gene level by a transcriptomic analysis using the massive RNA sequencing (RNA-seq) technology. Specifically, we have focused on differentially expressed genes that are related to neuronal and glial differentiation processes, as well as on groups of differentially expressed genes associated with different signaling pathways, in order to find a possible interaction between them and APP. Our data indicate a differential expression in genes related to Notch, Wnt, PI3K-AKT, and JAK-STAT signaling, among others. Knowledge of APP biological functions, as well as the possible signaling pathways that could be related to this protein, are essential to advance our understanding of AD.


Assuntos
Doença de Alzheimer , Células-Tronco Neurais , Humanos , Precursor de Proteína beta-Amiloide/genética , Fosfatidilinositol 3-Quinases , Neurogênese/genética , Doença de Alzheimer/genética , Transdução de Sinais
13.
Int J Mol Sci ; 24(19)2023 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-37834082

RESUMO

Amyloid precursor protein (APP) has been widely studied due to its association with Alzheimer's disease (AD). However, the physiological functions of APP are still largely unexplored. APP is a transmembrane glycoprotein whose expression in humans is abundant in the central nervous system. Specifically, several studies have revealed the high expression of APP during brain development. Previous studies in our laboratory revealed that a transient increase in APP expression induces early cell cycle exit of human neural stem cells (hNSCs) and directs their differentiation towards glial cells (gliogenesis) while decreasing their differentiation towards neurons (neurogenesis). In the present study, we have evaluated the intrinsic cellular effects of APP down-expression (using siRNA) on cell death, cell proliferation, and cell fate specification of hNSCs. Our data indicate that APP silencing causes cellular effects opposite to those obtained in previous APP overexpression assays, inducing cell proliferation in hNS1 cells (a model line of hNSCs) and favoring neurogenesis instead of gliogenesis in these cells. In addition, we have analyzed the gene and protein expression levels of ß-Catenin as a possible molecule involved in these cellular effects. These data could help to understand the biological role of APP, which is necessary to deepen the knowledge of AD.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Neurogênese , Humanos , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Células-Tronco Neurais/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo
14.
Int J Mol Sci ; 24(3)2023 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-36768434

RESUMO

Epigenetic regulation via epigenetic factors in collaboration with tissue-specific transcription factors is curtail for establishing functional organ systems during development. Brain development is tightly regulated by epigenetic factors, which are coordinately activated or inactivated during processes, and their dysregulation is linked to brain abnormalities and intellectual disability. However, the precise mechanism of epigenetic regulation in brain development and neurogenesis remains largely unknown. Here, we show that Tip60/KAT5 deletion in neural stem/progenitor cells (NSCs) in mice results in multiple abnormalities of brain development. Tip60-deficient embryonic brain led to microcephaly, and proliferating cells in the developing brain were reduced by Tip60 deficiency. In addition, neural differentiation and neuronal migration were severely affected in Tip60-deficient brains. Following neurogenesis in developing brains, gliogenesis started from the earlier stage of development in Tip60-deficient brains, indicating that Tip60 is involved in switching from neurogenesis to gliogenesis during brain development. It was also confirmed in vitro that poor neurosphere formation, proliferation defects, neural differentiation defects, and accelerated astrocytic differentiation in mutant NSCs are derived from Tip60-deficient embryonic brains. This study uncovers the critical role of Tip60 in brain development and NSC maintenance and function in vivo and in vitro.


Assuntos
Histona Acetiltransferases , Células-Tronco Neurais , Camundongos , Animais , Histona Acetiltransferases/genética , Epigênese Genética , Neurogênese , Células-Tronco Embrionárias , Diferenciação Celular/fisiologia
15.
J Neurosci Res ; 100(2): 578-597, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34811802

RESUMO

Traumatic brain injury (TBI) is a significant problem that affects over 800,000 children each year. As cell proliferation is disturbed by injury and required for normal brain development, we investigated how a pediatric closed head injury (CHI) would affect the progenitors of the subventricular zone (SVZ). Additionally, we evaluated the contribution of leukemia inhibitory factor (LIF) using germline LIF heterozygous mice (LIF Het), as LIF is an injury-induced cytokine, known to influence neurogenesis and gliogenesis. CHIs were performed on P20 LIF Het and wild-type (WT) mice. Ki-67 immunostaining and stereology revealed that cell proliferation increased ~250% in injured LIF Het mice compared to the 30% increase observed in injured WT mice at 48-hr post-CHI. OLIG2+ cell proliferation increased in the SVZ and white matter of LIF Het injured mice at 48-hr recovery. Using an 8-color flow cytometry panel, the proliferation of three distinct multipotential progenitors and early oligodendrocyte progenitor cell proliferation was significantly increased in LIF Het injured mice compared to WT injured mice. Supporting its cytostatic function, LIF decreased neurosphere progenitor and oligodendrocyte progenitor cell proliferation compared to controls. In highly enriched mouse oligodendrocyte progenitor cell cultures, LIF increased phospho-protein kinase B after 20 min and increased phospho-S6 ribosomal protein at 20 and 40 min of exposure, which are downstream targets of the mammalian target of rapamycin pathway. Altogether, our data provide new insights into the regulatory role of LIF in suppressing neural progenitor cell proliferation and, in particular, oligodendrocyte progenitor cell proliferation after a mild TBI.


Assuntos
Lesões Encefálicas Traumáticas , Células Precursoras de Oligodendrócitos , Animais , Proliferação de Células/fisiologia , Humanos , Fator Inibidor de Leucemia , Mamíferos , Camundongos , Camundongos Endogâmicos C57BL , Neurogênese/fisiologia
16.
Cell Mol Neurobiol ; 42(6): 1859-1873, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33666795

RESUMO

Ischemic preconditioning (IPC) is an approach of protection against cerebral ischemia by inducing endogenous cytoprotective machinery. However, few studies in neurogenesis and oligodendrogenesis after IPC have been reported, especially the latter. The purpose of this study is to test our hypothesis that IPC may also induce cell proliferation and oligodendrogenesis in the subventricular zone and striatum, as well as to investigate the effect of nuclear factor erythroid 2-related factor 2 (Nrf2) on oligodendrogenesis. IPC was induced in mice by 12-min ischemia through the occlusion of the middle cerebral artery. Newly generated cells were labeled with 5-bromo-2'-deoxyuridine. Our findings demonstrated that IPC stimulated the proliferation of neural stem cells in the subventricular zone, promoted the generation of oligodendrocyte precursor cells in the striatum and corpus callosum/external capsule (CC/EC), and stimulated oligodendrocyte precursor cells differentiation into oligodendrocytes in the striatum and the CC/EC. Furthermore, we describe a crucial role for Nrf2 in IPC-induced oligodendrogenesis in the subventricular zone, striatum, and CC/EC and show for the first time that Nrf2 promoted the migration and differentiation of oligodendrocyte precursor cells into oligodendrocytes in the striatum and CC/EC. Our data imply that IPC stimulates the oligodendrogenesis in the brain and that Nrf2 signaling may contribute to the oligodendrogenesis.


Assuntos
Precondicionamento Isquêmico , Fator 2 Relacionado a NF-E2 , Células Precursoras de Oligodendrócitos , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Bromodesoxiuridina , Camundongos , Fator 2 Relacionado a NF-E2/deficiência , Fator 2 Relacionado a NF-E2/fisiologia , Células Precursoras de Oligodendrócitos/citologia , Oligodendroglia/citologia
17.
World J Surg Oncol ; 20(1): 146, 2022 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-35538578

RESUMO

BACKGROUND: Glioblastoma is one of the most aggressive tumors. The etiology and the factors determining its onset are not yet entirely known. This study investigates the origins of GBM, and for this purpose, it focuses primarily on developmental gliogenic processes. It also focuses on the impact of the related neurogenic developmental processes in glioblastoma oncogenesis. It also addresses why glial cells are at more risk of tumor development compared to neurons. METHODS: Databases including PubMed, MEDLINE, and Google Scholar were searched for published articles without any date restrictions, involving glioblastoma, gliogenesis, neurogenesis, stemness, neural stem cells, gliogenic signaling and pathways, neurogenic signaling and pathways, and astrocytogenic genes. RESULTS: The origin of GBM is dependent on dysregulation in multiple genes and pathways that accumulatively converge the cells towards oncogenesis. There are multiple layers of steps in glioblastoma oncogenesis including the failure of cell fate-specific genes to keep the cells differentiated in their specific cell types such as p300, BMP, HOPX, and NRSF/REST. There are genes and signaling pathways that are involved in differentiation and also contribute to GBM such as FGFR3, JAK-STAT, and hey1. The genes that contribute to differentiation processes but also contribute to stemness in GBM include notch, Sox9, Sox4, c-myc gene overrides p300, and then GFAP, leading to upregulation of nestin, SHH, NF-κB, and others. GBM mutations pathologically impact the cell circuitry such as the interaction between Sox2 and JAK-STAT pathway, resulting in GBM development and progression. CONCLUSION: Glioblastoma originates when the gene expression of key gliogenic genes and signaling pathways become dysregulated. This study identifies key gliogenic genes having the ability to control oncogenesis in glioblastoma cells, including p300, BMP, PAX6, HOPX, NRSF/REST, LIF, and TGF beta. It also identifies key neurogenic genes having the ability to control oncogenesis including PAX6, neurogenins including Ngn1, NeuroD1, NeuroD4, Numb, NKX6-1 Ebf, Myt1, and ASCL1. This study also postulates how aging contributes to the onset of glioblastoma by dysregulating the gene expression of NF-κB, REST/NRSF, ERK, AKT, EGFR, and others.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Neoplasias Encefálicas/patologia , Carcinogênese/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Glioblastoma/patologia , Humanos , Janus Quinases/genética , Janus Quinases/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Células-Tronco Neoplásicas/metabolismo , Neurogênese , Fatores de Transcrição SOXC/genética , Fatores de Transcrição STAT/genética , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais
18.
Int J Mol Sci ; 23(18)2022 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-36142348

RESUMO

Aquaporin 4 (AQP4) is a cerebral glial marker that labels ependymal cells and astrocytes' endfeet and is the main water channel responsible for the parenchymal fluid balance. However, in brain development, AQP4 is a marker of glial stem cells and plays a crucial role in the pathophysiology of pediatric hydrocephalus. Gliogenesis characterization has been hampered by a lack of biomarkers for precursor and intermediate stages and a deeper understanding of hydrocephalus etiology is needed. This manuscript is a focused review of the current research landscape on AQP4 as a possible biomarker for gliogenesis and its influence in pediatric hydrocephalus, emphasizing reactive astrogliosis. The goal is to understand brain development under hydrocephalic and normal physiologic conditions.


Assuntos
Aquaporina 4 , Hidrocefalia , Astrócitos/metabolismo , Criança , Gliose , Humanos , Neuroglia/metabolismo , Água/metabolismo
19.
J Neurosci ; 40(7): 1501-1513, 2020 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-31949107

RESUMO

The bHLH transcription factor Hes1 is a key downstream effector for the Notch signaling pathway. During embryogenesis neural progenitors express low levels of Hes1 in an oscillating pattern, whereas glial brain boundary regions (e.g., isthmus) have high, sustained Hes1 levels that suppress neuronal fates. Here, we show that in the embryonic mouse retina, the optic nerve head and stalk express high Hes1, with the ONH constituting a boundary between the neural retina and glial cells that ultimately line the optic stalk. Using two Cre drivers with distinct spatiotemporal expression we conditionally inactivated Hes1, to delineate the requirements for this transcriptional repressor during retinal neurogenesis versus patterning of the optic cup and stalk. Throughout retinal neurogenesis, Hes1 maintains proliferation and blocks retinal ganglion cell formation, but surprisingly we found it also promotes cone photoreceptor genesis. In the postnatal eye, Hes1 inactivation with Rax-Cre resulted in increased bipolar neurons and a mispositioning of Müller glia. Our results indicate that Notch pathway regulation of cone genesis is more complex than previously assumed, and reveal a novel role for Hes1 in maintaining the optic cup-stalk boundary.SIGNIFICANCE STATEMENT The bHLH repressor Hes1 regulates the timing of neurogenesis, rate of progenitor cell division, gliogenesis, and maintains tissue compartment boundaries. This study expands current eye development models by showing Notch-independent roles for Hes1 in the developing optic nerve head (ONH). Defects in ONH formation result in optic nerve coloboma; our work now inserts Hes1 into the genetic hierarchy regulating optic fissure closure. Given that Hes1 acts analogously in the ONH as the brain isthmus, it prompts future investigation of the ONH as a signaling factor center, or local organizer. Embryonic development of the ONH region has been poorly studied, which is surprising given it is where the pan-ocular disease glaucoma is widely believed to inflict damage on RGC axons.


Assuntos
Olho/embriologia , Neurogênese/fisiologia , Fatores de Transcrição HES-1/fisiologia , Animais , Coloboma/genética , Coloboma/patologia , Células Ependimogliais/citologia , Olho/crescimento & desenvolvimento , Gastrulação , Estudos de Associação Genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microftalmia/genética , Microftalmia/patologia , Disco Óptico/embriologia , Disco Óptico/patologia , Receptores Notch/fisiologia , Retina/anormalidades , Retina/embriologia , Células Bipolares da Retina/citologia , Células Fotorreceptoras Retinianas Cones/metabolismo , Células Ganglionares da Retina/citologia , Transdução de Sinais , Fatores de Transcrição HES-1/deficiência , Fatores de Transcrição HES-1/genética
20.
Semin Cell Dev Biol ; 95: 4-11, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30634047

RESUMO

In the developing mammalian neocortex, neural stem cells (NSCs) gradually alter their characteristics as development proceeds. NSCs initially expand the progenitor pool by symmetric proliferative division and then shift to asymmetric neurogenic division to commence neurogenesis. NSCs sequentially give rise to deep layer neurons first and superficial layer neurons later through mid- to late-embryonic stages, followed by shifting to a gliogenic phase at perinatal stages. The precise mechanisms regulating developmental timing of the transition from symmetric to asymmetric division have not been fully elucidated; however, gradual elongation in cell cycle length and concomitant accumulation of determinants that promote neuronal differentiation may function as a biological clock that regulates the onset of asymmetric neurogenic division. On the other hand, epigenetic regulatory systems have been implicated in the regulation of transition timing of neurogenesis and gliogenesis; the polycomb group (PcG) complex and Hmga genes have been found to govern the developmental timing by modulating chromatin structure during neocortical development. Furthermore, we uncovered several factors and mechanisms underlying the regulation of timing of neocortical neurogenesis and gliogenesis. In this review, we discuss recent findings regarding the mechanisms that govern the temporal properties of NSCs and the precise transition timing during neocortical development.


Assuntos
Mamíferos/embriologia , Neocórtex/embriologia , Células-Tronco Neurais/citologia , Neurogênese , Neuroglia/citologia , Animais , Humanos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA