Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mov Disord ; 39(3): 498-509, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38148610

RESUMO

BACKGROUND: Kinase D-interacting substrate of 220 kDa (KIDINS220) is a multifunctional scaffolding protein essential for neuronal development. It has been implicated in neurological diseases with either autosomal dominant (AD) or autosomal recessive (AR) inheritance patterns. The molecular mechanisms underlying the AR/AD dual nature of KIDINS220 remain elusive, posing challenges to genetic interpretation and clinical interventions. Moreover, increased KIDINS220 exhibited neurotoxicity, but its role in neurodevelopment remains unclear. OBJECTIVE: The aim was to investigate the genotype-phenotype correlations of KIDINS220 and elucidate its pathophysiological role in neuronal development. METHODS: Whole-exome sequencing was performed in a four-generation family with cerebral palsy. CRISPR/Cas9 was used to generate KIDINS220 mutant cell lines. In utero electroporation was employed to investigate the effect of KIDINS220 variants on neurogenesis in vivo. RESULTS: We identified in KIDINS220 a pathogenic nonsense variant (c.4177C > T, p.Q1393*) that associated with AD cerebral palsy. We demonstrated that the nonsense variants located in the terminal exon of KIDINS220 are gain-of-function (GoF) variants, which enable the mRNA to escape nonsense-mediated decay and produce a truncated yet functional KIDINS220 protein. The truncated protein exhibited significant resistance to calpain and consequently accumulated within cells, resulting in the hyperactivation of Rac1 and defects in neuronal development. CONCLUSIONS: Our findings demonstrate that the location of variants within KIDINS220 plays a crucial role in determining inheritance patterns and corresponding clinical outcomes. The proposed interaction between Rac1 and KIDINS220 provides new insights into the pathogenesis of cerebral palsy, implying potential therapeutic perspectives. © 2023 International Parkinson and Movement Disorder Society.


Assuntos
Paralisia Cerebral , Neurônios , Humanos , Neurônios/metabolismo , Transdução de Sinais , Paralisia Cerebral/genética , Mutação com Ganho de Função , Neurogênese/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/genética
2.
BMC Med ; 21(1): 510, 2023 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-38129829

RESUMO

BACKGROUND: Exposure to general anesthesia influences neuronal functions during brain development. Recently, interneurons were found to be involved in developmental neurotoxicity by anesthetic exposure. But the underlying mechanism and long-term consequences remain elusive. METHODS: Pregnant mice received 2.5% sevoflurane for 6-h on gestational day 14.5. Pentylenetetrazole (PTZ)-induced seizure, anxiety- and depression-like behavior tests were performed in 30- and 60-day-old male offspring. Cortical interneurons were labeled using Rosa26-EYFP/-; Nkx2.1-Cre mice. Immunofluorescence and electrophysiology were performed to determine the cortical interneuron properties. Q-PCR and in situ hybridization (ISH) were performed for the potential mechanism, and the finding was further validated by in utero electroporation (IUE). RESULTS: In this study, we found that maternal sevoflurane exposure increased epilepsy susceptibility by using pentylenetetrazole (PTZ) induced-kindling models and enhanced anxiety- and depression-like behaviors in adolescent offspring. After sevoflurane exposure, the highly ordered cortical interneuron migration was disrupted in the fetal cortex. In addition, the resting membrane potentials of fast-spiking interneurons in the sevoflurane-treated group were more hyperpolarized in adolescence accompanied by an increase in inhibitory synapses. Both q-PCR and ISH indicated that CXCL12/CXCR4 signaling pathway downregulation might be a potential mechanism under sevoflurane developmental neurotoxicity which was further confirmed by IUE and behavioral tests. Although the above effects were obvious in adolescence, they did not persist into adulthood. CONCLUSIONS: Our findings demonstrate that maternal anesthesia impairs interneuron migration through the CXCL12/CXCR4 signaling pathway, and influences the interneuron properties, leading to the increased epilepsy susceptibility in adolescent offspring. Our study provides a novel perspective on the developmental neurotoxicity of the mechanistic link between maternal use of general anesthesia and increased susceptibility to epilepsy.


Assuntos
Epilepsia , Pentilenotetrazol , Humanos , Gravidez , Feminino , Camundongos , Animais , Masculino , Sevoflurano/metabolismo , Sevoflurano/farmacologia , Pentilenotetrazol/toxicidade , Pentilenotetrazol/metabolismo , Exposição Materna/efeitos adversos , Interneurônios/metabolismo , Epilepsia/induzido quimicamente
3.
Cell Rep ; 42(7): 112677, 2023 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-37352102

RESUMO

Polarity proteins regulate the proliferation and differentiation of neural progenitors to generate neurons during brain development through multiple signaling pathways. However, how cell polarity couples the signaling pathways remains unclear. Here, we show that coiled-coil domain-containing protein 85c (Ccdc85c) interacts with the polarity protein Par3 to regulate the proliferation of radial glial cells (RGCs) via phase separation coupled to percolation (PSCP). We find that the interaction with Ccdc85c relieves the intramolecular auto-inhibition of Par3, which leads to PSCP of Par3. Downregulation of Ccdc85c causes RGC differentiation. Importantly, the open conformation of Par3 facilitates the recruitment of the Notch regulator Numb to the Par3 condensates, which might prevent the attenuation of Notch activity to maintain RGC proliferation. Furthermore, ectopic activation of Notch signaling rescues RGC proliferation defects caused by the downregulation of Ccdc85c. These results suggest that Ccdc85c-mediated PSCP of Par3 regulates Notch signaling to control RGC proliferation during brain development.


Assuntos
Polaridade Celular , Transdução de Sinais , Polaridade Celular/fisiologia , Transdução de Sinais/fisiologia , Neurônios/metabolismo , Proliferação de Células , Receptores Notch/metabolismo
5.
Cell Prolif ; 54(6): e13042, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33955094

RESUMO

OBJECTIVES: The effects of general anaesthetics on fetal brain development remain elusive. Radial glial progenitors (RGPs) generate the majority of neurons in developing brains. Here, we evaluated the acute alterations in RGPs after maternal sevoflurane exposure. METHODS: Pregnant mice were exposed to 2.5% sevoflurane for 6 hours on gestational day 14.5. Interkinetic nuclear migration (INM) of RGPs in the ventricular zone (VZ) of the fetal brain was evaluated by thymidine analogues labelling. Cell fate of RGP progeny was determined by immunostaining using various neural markers. The Morris water maze (MWM) was used to assess the neurocognitive behaviours of the offspring. RNA sequencing (RNA-Seq) was performed for the potential mechanism, and the potential mechanism validated by quantitative real-time PCR (qPCR), Western blot and rescue experiments. Furthermore, INM was examined in human embryonic stem cell (hESC)-derived 3D cerebral organoids. RESULTS: Maternal sevoflurane exposure induced temporary abnormities in INM, and disturbed the cell cycle progression of RGPs in both rodents and cerebral organoids without cell fate alternation. RNA-Seq analysis, qPCR and Western blot showed that the Notch signalling pathway was a potential downstream target. Reactivation of Notch by Jag1 and NICD overexpression rescued the defects in INM. Young adult offspring showed no obvious cognitive impairments in MWM. CONCLUSIONS: Maternal sevoflurane exposure during neurogenic period temporarily induced abnormal INM of RGPs by targeting the Notch signalling pathway without inducing long-term effects on RGP progeny cell fate or offspring cognitive behaviours. More importantly, the defects of INM in hESC-derived cerebral organoids provide a novel insight into the effects of general anaesthesia on human brain development.


Assuntos
Anestésicos Inalatórios/efeitos adversos , Córtex Cerebral/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Receptores Notch/metabolismo , Sevoflurano/efeitos adversos , Animais , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Córtex Cerebral/patologia , Feminino , Feto/efeitos dos fármacos , Feto/metabolismo , Feto/patologia , Humanos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/citologia , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/patologia , Neurogênese/efeitos dos fármacos , Neuroglia/citologia , Neuroglia/efeitos dos fármacos , Neuroglia/patologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/patologia , Transdução de Sinais/efeitos dos fármacos
6.
Mol Psychiatry ; 26(11): 6562-6577, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33963287

RESUMO

The establishment of neural circuits depends on precise neuronal positioning in the cortex, which occurs via a tightly coordinated process of neuronal differentiation, migration, and terminal localization. Deficits in this process have been implicated in several psychiatric disorders. Here, we show that the transcription factor Tcf4 controls neuronal positioning during brain development. Tcf4-deficient neurons become mispositioned in clusters when their migration to the cortical plate is complete. We reveal that Tcf4 regulates the expression of cell adhesion molecules to control neuronal positioning. Furthermore, through in vivo extracellular electrophysiology, we show that neuronal functions are disrupted after the loss of Tcf4. TCF4 mutations are strongly associated with schizophrenia and cause Pitt-Hopkins syndrome, which is characterized by severe intellectual disability. Thus, our results not only reveal the importance of neuronal positioning in brain development but also provide new insights into the potential mechanisms underlying neurological defects linked to TCF4 mutations.


Assuntos
Hiperventilação , Deficiência Intelectual , Adesão Celular , Fácies , Humanos , Hiperventilação/genética , Hiperventilação/metabolismo , Deficiência Intelectual/genética , Neurônios/metabolismo , Fator de Transcrição 4/genética , Fator de Transcrição 4/metabolismo
7.
G3 (Bethesda) ; 11(8)2021 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-33744924

RESUMO

Intellectual disability is closely related to impaired GABA neurotransmission. Brpf1 was specifically expressed in medial ganglionic eminence (MGE), a developmental niche of GABAergic interneurons, and patients with BRPF1 mutations showed intellectual disability. To test its role in the development and function of MGE-derived GABAergic interneurons, we performed immunofluorescence staining, whole-cell patch-clamp, MGE transplantation, and mRNA-Seq to understand its effect on neuronal differentiation, dendritic morphology, electrophysiology, migration, and gene regulation, using mouse MGE-derived GABAergic interneurons infected with AAV-shBrpf1. The results showed that Brpf1 knockdown had a decreasing trend, although not significant, on the differentiation of GABAergic interneurons into parvalbumin+ interneurons. Moreover, increased firing threshold, decreased number of evoked action potentials, and a reduced amplitude of miniature inhibitory postsynaptic currents were observed before any significant change of MAP2+ dendritic morphology and in vivo migration ability appeared. Finally, mRNA-Seq analysis revealed that genes related to neurodevelopment and synaptic transmission such as Map2k7 were dysregulated. Our results demonstrated a key role of Brpf1 in inhibitory neurotransmission and related gene expression of GABAergic interneurons.


Assuntos
Deficiência Intelectual , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Ligação a DNA , Humanos , Deficiência Intelectual/genética , Interneurônios , Eminência Mediana , Camundongos , Transmissão Sináptica
8.
Cell Death Dis ; 12(1): 91, 2021 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-33462220

RESUMO

A proper number of oligodendrocytes in the nerve system is essential for neuronal functions. In the olfactory bulb (OB), enriched oligodendrocytes are crucial for olfactory information processing. However, how the precise number of oligodendrocytes in the OB is regulated remains elusive. Here we identified that the transcription factor 4 (Tcf4)-mediated cell death is essential for generating an appropriate number of oligodendrocyte progenitor cells (OPCs) and thereby oligodendrocytes in the OB. We showed that Nkx2.1-positive progenitors in the medial ganglionic eminence (MGE) and anterior entopeduncular area (AEP) provide the first source of OPCs in the OB. Conditional depletion of Tcf4 leads to an increase of OPCs in the OB, which is mediated by the suppression of programmed cell death. Furthermore, we showed that Tcf4 mediated OPC survival is cell-autonomous by transplantation assay. Mechanistically, we identified Bax/Bak as a potential key pathway to promote OPC elimination during OB development. Depletion of Bax/Bak in Nkx2.1 lineage results in an increase of OPCs in the OB. Mutations in TCF4 causes Pitt-Hopkins syndrome, a severe neurodevelopmental disorder. Thus, our findings reveal an important intrinsic mechanism underlying the survival control of OPCs in the OB and provide new insights into the pathogenesis of Pitt-Hopkins syndrome.


Assuntos
Bulbo Olfatório/metabolismo , Células Precursoras de Oligodendrócitos/metabolismo , Oligodendroglia/metabolismo , Fator de Transcrição 4/metabolismo , Animais , Sobrevivência Celular/fisiologia , Humanos , Camundongos , Camundongos Knockout , Neurogênese , Bulbo Olfatório/citologia , Bulbo Olfatório/crescimento & desenvolvimento , Células Precursoras de Oligodendrócitos/citologia , Oligodendroglia/citologia
9.
Cereb Cortex ; 30(5): 3102-3115, 2020 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-31845732

RESUMO

The dentate gyrus (DG) of the hippocampal formation plays essential roles in learning and memory. Defective DG development is associated with neurological disorders. Here, we show that transcription factor 4 (Tcf4) is essential for DG development. Tcf4 expression is elevated in neural progenitors of the dentate neuroepithelium in the developing mouse brain. We demonstrate that conditional disruption of Tcf4 in the dentate neuroepithelium leads to abnormal neural progenitor migration guided by disorganized radial glial fibers, which further leads to hypoplasia in the DG. Moreover, we reveal that Wnt7b is a key downstream effector of Tcf4 in regulating neural progenitor migration. Behavioral analysis shows that disruption of integrity of the DG impairs the social memory highlighting the importance of proper development of the DG. These results reveal a critical role for Tcf4 in regulating DG development. As mutations in TCF4 cause Pitt-Hopkins syndrome (PTHS) characterized by severe intellectual disability, our data also potentially provide insights into the basis of neurological defects linked to TCF4 mutations.


Assuntos
Movimento Celular/fisiologia , Giro Denteado/crescimento & desenvolvimento , Giro Denteado/metabolismo , Células-Tronco Neurais/metabolismo , Fator de Transcrição 4/biossíntese , Animais , Giro Denteado/embriologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Gravidez , Fator de Transcrição 4/genética
10.
Cell Death Differ ; 26(11): 2479-2492, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30858607

RESUMO

During neocortical development, there are two important events, including expansion of the neural progenitor pool through symmetric divisions, and generation of neurons via asymmetrical divisions that lead to a serial process of neuronal polarization, migration, and layer-type specific phenotype acquisition. The mechanisms underlying these processes remain poorly elucidated. Here, we show that the transcription factor Zeb1 regulates the orientation of the cleavage plane of dividing neural progenitors, neuronal polarity, and migration. Upon Zeb1 removal, the cleavage plane of mitotic neural progenitors fails to orientate vertically, resulting in random orientation and premature neuronal differentiation. Consequently, these extra number of precociously produced neurons migrate aberrantly to the upper layer. Mechanistically, we show that Zeb1 suppresses Pak3, a p21-activated serine/threonine protein kinase, through formation of a functional repressing complex together with methyltransferase PRMT5 and Pak3. Our results reveal that Zeb1 plays an essential role in neocortical development and may provide insights into the mechanisms responsible for cortical developmental diseases.


Assuntos
Células Ependimogliais/citologia , Neocórtex/embriologia , Células-Tronco Neurais/citologia , Neurogênese/fisiologia , Neurônios/citologia , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo , Animais , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Camundongos , Camundongos Knockout , Regiões Promotoras Genéticas/genética , Proteína-Arginina N-Metiltransferases/metabolismo , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética , Quinases Ativadas por p21/metabolismo
11.
Neuron ; 101(6): 1117-1133.e5, 2019 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-30709655

RESUMO

Neural progenitors with distinct potential to generate progeny are associated with a spatially distinct microenvironment. Neocortical intermediate progenitors (IPs) in the subventricular zone (SVZ) of the developing brain generate neurons for all cortical layers and are essential for cortical expansion. Here, we show that spatial control of IP positioning is essential for neocortical development. We demonstrate that HDAC1 and HDAC2 regulate the spatial positioning of IPs to form the SVZ. Developmental stage-specific depletion of both HDAC1 and HDAC2 in radial glial progenitors results in mispositioning of IPs at the ventricular surface, where they divide and differentiate into neurons, thereby leading to the cortical malformation. We further identified the proneural gene Neurogenin2 as a key target of HDAC1 and HDAC2 for regulating IP positioning. Our results demonstrate the importance of the spatial positioning of neural progenitors in cortical development and reveal a mechanism underlying the establishment of the SVZ microenvironment.


Assuntos
Células Ependimogliais/metabolismo , Histona Desacetilase 1/genética , Histona Desacetilase 2/genética , Ventrículos Laterais/embriologia , Malformações do Desenvolvimento Cortical/genética , Neocórtex/embriologia , Células-Tronco Neurais/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Movimento Celular/genética , Histona Desacetilase 1/metabolismo , Histona Desacetilase 2/metabolismo , Malformações do Desenvolvimento Cortical/embriologia , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Neurogênese
12.
Toxicol Sci ; 160(2): 268-283, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-28973681

RESUMO

Lead (Pb) is a toxic heavy metal affecting human health; it is known to be harmful to various organs or systems, yet the mechanisms by which Pb influences immune cell development remain to be defined. In this study, we show that Pb exposure (1250 ppm via drinking water) selectively impacted the development of myeloid cells (myelopoiesis). After Pb treatment of adult C57BL/6 mice, the numbers of granulocyte-macrophage progenitors (GMP) were consistently reduced, whereas the numbers of myeloid cells were increased at week (wk) 1 and decreased at wk8 after initiating the Pb exposure. Functional assays indicate that Pb accelerated GMP differentiation in a reactive oxygen species-dependent manner after treatment for 1 week and inhibited common myeloid progenitor differentiation by upregulating interferon regulatory factor 8 (IRF8) expression after treatment for 8 weeks. Consistent with the distinct Pb influences on myeloid cells observed at wk1 and wk8, Pb caused an inflammatory environment in vivo at wk8, but not at wk1. Furthermore, like the observations in mice during the Pb exposure, bloods from humans occupationally exposed to Pb had their numbers of monocytes, neutrophils and GMP negatively associated with the Pb concentration, whereas IRF8 expression in common myeloid progenitor, but not GMP, was positively correlated with the Pb concentration. These data suggest an occupationally relevant level of Pb exposure preferentially influences myelopoiesis involving reactive oxygen species and IRF8, which may contribute to the current understanding of the hematopoietic toxicology of Pb.


Assuntos
Linhagem da Célula/efeitos dos fármacos , Poluentes Ambientais/efeitos adversos , Células Progenitoras de Granulócitos e Macrófagos/efeitos dos fármacos , Células Progenitoras Mieloides/efeitos dos fármacos , Mielopoese/efeitos dos fármacos , Exposição Ocupacional/efeitos adversos , Compostos Organometálicos/efeitos adversos , Animais , Células Cultivadas , Técnicas de Cocultura , Poluentes Ambientais/sangue , Feminino , Células Progenitoras de Granulócitos e Macrófagos/metabolismo , Células Progenitoras de Granulócitos e Macrófagos/patologia , Humanos , Fatores Reguladores de Interferon/metabolismo , Contagem de Leucócitos , Masculino , Camundongos Endogâmicos C57BL , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/patologia , Compostos Organometálicos/sangue , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
13.
EMBO Rep ; 18(9): 1618-1630, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28747490

RESUMO

The Nedd4 family E3 ligases are key regulators of cell growth and proliferation and are often misregulated in human cancers and other diseases. The ligase activities of Nedd4 E3s are tightly controlled via auto-inhibition. However, the molecular mechanism underlying Nedd4 E3 auto-inhibition and activation is poorly understood. Here, we show that the WW domains proceeding the catalytic HECT domain play an inhibitory role by binding directly to HECT in the Nedd4 E3 family member Itch. Our structural and biochemical analyses of Itch reveal that the WW2 domain and a following linker allosterically lock HECT in an inactive state inhibiting E2-E3 transthiolation. Binding of the Ndfip1 adaptor or JNK1-mediated phosphorylation relieves the auto-inhibition of Itch in a WW2-dependent manner. Aberrant activation of Itch leads to migration defects of cortical neurons during development. Our study provides a new mechanism governing the regulation of Itch.


Assuntos
Ubiquitina-Proteína Ligases Nedd4/química , Ubiquitina-Proteína Ligases Nedd4/metabolismo , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/metabolismo , Regulação Alostérica , Animais , Cristalografia por Raios X , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Humanos , Camundongos , Ubiquitina-Proteína Ligases Nedd4/genética , Fosforilação , Ligação Proteica , Estrutura Terciária de Proteína , Proteólise , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Ubiquitina-Proteína Ligases/genética , Ubiquitinação , Domínios WW
14.
Toxicol Appl Pharmacol ; 313: 24-34, 2016 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-27771405

RESUMO

The heavy metal cadmium (Cd) is known to modulate immunity and cause osteoporosis. However, how Cd influences on hematopoiesis remain largely unknown. Herein, we show that wild-type C57BL/6 (B6) mice exposed to Cd for 3months had expanded bone marrow (BM) populations of long-term hematopoietic stem cells (LT-HSCs), common myeloid progenitors (CMPs) and granulocyte-macrophage progenitors (GMPs), while having reduced populations of multipotent progenitors (MPPs) and common lymphoid progenitors (CLPs). A competitive mixed BM transplantation assay indicates that BM from Cd-treated mice had impaired LT-HSC ability to differentiate into mature cells. In accordance with increased myeloid progenitors and decreased lymphoid progenitors, the BM and spleens of Cd-treated mice had more monocytes and/or neutrophils and fewer B cells and T cells. Cd impaired the ability of the non-hematopoietic system to support LT-HSCs, in that lethally irradiated Cd-treated recipients transplanted with normal BM cells had reduced LT-HSCs after the hematopoietic system was fully reconstituted. This is consistent with reduced osteoblasts, a known critical component for HSC niche, observed in Cd-treated mice. Conversely, lethally irradiated control recipients transplanted with BM cells from Cd-treated mice had normal LT-HSC reconstitution. Furthermore, both control mice and Cd-treated mice that received Alendronate, a clinical drug used for treating osteoporosis, had BM increases of LT-HSCs. Thus, the results suggest Cd increase of LT-HSCs is due to effects on HSCs and not on osteoblasts, although, Cd causes osteoblast reduction and impaired niche function for maintaining HSCs. Furthermore, Cd skews HSCs toward myelopoiesis.


Assuntos
Cádmio/toxicidade , Células-Tronco Hematopoéticas/efeitos dos fármacos , Mielopoese/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Alendronato/farmacologia , Animais , Carga Corporal (Radioterapia) , Osso e Ossos/citologia , Osso e Ossos/efeitos dos fármacos , Cádmio/farmacocinética , Camundongos , Camundongos Endogâmicos C57BL , Osteoblastos/efeitos dos fármacos
15.
Development ; 141(3): 604-616, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24449838

RESUMO

The histone deacetylases HDAC1 and HDAC2 are crucial regulators of chromatin structure and gene expression, thereby controlling important developmental processes. In the mouse brain, HDAC1 and HDAC2 exhibit different developmental stage- and lineage-specific expression patterns. To examine the individual contribution of these deacetylases during brain development, we deleted different combinations of Hdac1 and Hdac2 alleles in neural cells. Ablation of Hdac1 or Hdac2 by Nestin-Cre had no obvious consequences on brain development and architecture owing to compensation by the paralog. By contrast, combined deletion of Hdac1 and Hdac2 resulted in impaired chromatin structure, DNA damage, apoptosis and embryonic lethality. To dissect the individual roles of HDAC1 and HDAC2, we expressed single alleles of either Hdac1 or Hdac2 in the absence of the respective paralog in neural cells. The DNA-damage phenotype observed in double knockout brains was prevented by expression of a single allele of either Hdac1 or Hdac2. Strikingly, Hdac1(-/-)Hdac2(+/-) brains showed normal development and no obvious phenotype, whereas Hdac1(+/-)Hdac2(-/-) mice displayed impaired brain development and perinatal lethality. Hdac1(+/-)Hdac2(-/-) neural precursor cells showed reduced proliferation and premature differentiation mediated by overexpression of protein kinase C, delta, which is a direct target of HDAC2. Importantly, chemical inhibition or knockdown of protein kinase C delta was sufficient to rescue the phenotype of neural progenitor cells in vitro. Our data indicate that HDAC1 and HDAC2 have a common function in maintaining proper chromatin structures and show that HDAC2 has a unique role by controlling the fate of neural progenitors during normal brain development.


Assuntos
Alelos , Encéfalo/embriologia , Encéfalo/enzimologia , Histona Desacetilase 1/metabolismo , Histona Desacetilase 2/genética , Homologia de Sequência de Aminoácidos , Acetofenonas/farmacologia , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Apoptose/genética , Benzopiranos/farmacologia , Encéfalo/metabolismo , Encéfalo/patologia , Proteínas Correpressoras/metabolismo , Dano ao DNA/genética , Perda do Embrião/enzimologia , Perda do Embrião/patologia , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Histona Desacetilase 1/genética , Histona Desacetilase 2/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Proteína Quinase C-delta/antagonistas & inibidores , Proteína Quinase C-delta/genética , Proteína Quinase C-delta/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
16.
Proc Natl Acad Sci U S A ; 111(3): 1014-9, 2014 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-24381158

RESUMO

The orientation of the mitotic spindle determines the relative size and position of the daughter cells and influences the asymmetric inheritance of localized cell fate determinants. The onset of mammalian neurogenesis, for example, coincides with changes in spindle orientation. To address the functional implications of this and related phenomena, precise methods for determining the orientation of the mitotic spindle in complex tissues are needed. Here, we present methodology for the analysis of spindle orientation in 3D. Our method allows statistical analysis and modeling of spindle orientation and involves two parameters for horizontal and vertical bias that can unambiguously describe the distribution of spindle orientations in an experimental sample. We find that 3D analysis leads to systematically different results from 2D analysis and, surprisingly, truly random spindle orientations do not result in equal numbers of horizontal and vertical orientations. We show that our method can describe the distribution of spindle orientation angles under different biological conditions. As an example of biological application we demonstrate that the adapter protein Inscuteable (mInsc) can actively promote vertical spindle orientation in apical progenitors during mouse neurogenesis.


Assuntos
Fuso Acromático , Algoritmos , Animais , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular/genética , Divisão Celular , Linhagem da Célula , Polaridade Celular/genética , Simulação por Computador , Imageamento Tridimensional , Camundongos , Neurogênese/fisiologia , Neurônios/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Probabilidade , Processos Estocásticos
17.
Neuron ; 79(2): 254-65, 2013 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-23830831

RESUMO

In the developing neocortex, progenitor cells expand through symmetric division before they generate cortical neurons through multiple rounds of asymmetric cell division. Here, we show that the orientation of the mitotic spindle plays a crucial role in regulating the transition between those two division modes. We demonstrate that the protein phosphatase PP4c regulates spindle orientation in early cortical progenitor cells. Upon removing PP4c, mitotic spindles fail to orient in parallel to the neuroepithelial surface and progenitors divide with random orientation. As a result, their divisions become asymmetric and neurogenesis starts prematurely. Biochemical and genetic experiments show that PP4c acts by dephosphorylating the microtubule binding protein Ndel1, thereby enabling complex formation with Lis1 to form a functional spindle orientation complex. Our results identify a key regulator of cortical development and demonstrate that changes in the orientation of progenitor division are responsible for the transition between symmetric and asymmetric cell division.


Assuntos
Proliferação de Células , Neocórtex/embriologia , Neocórtex/enzimologia , Neurogênese/fisiologia , Fosfoproteínas Fosfatases/fisiologia , Fuso Acromático/enzimologia , Animais , Divisão Celular/fisiologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neocórtex/citologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/enzimologia , Gravidez
18.
EMBO J ; 32(11): 1598-612, 2013 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-23624931

RESUMO

Polycomb group (PcG) proteins form transcriptional repressor complexes with well-established functions during cell-fate determination. Yet, the mechanisms underlying their regulation remain poorly understood. Here, we extend the role of Polycomb complexes in the temporal control of neural progenitor cell (NPC) commitment by demonstrating that the PcG protein Ezh2 is necessary to prevent the premature onset of gliogenesis. In addition, we identify the chromodomain helicase DNA-binding protein 4 (Chd4) as a critical interaction partner of Ezh2 required specifically for PcG-mediated suppression of the key astrogenic marker gene GFAP. Accordingly, in vivo depletion of Chd4 in the developing neocortex promotes astrogenesis. Collectively, these results demonstrate that PcG proteins operate in a highly dynamic, developmental stage-dependent fashion during neural differentiation and suggest that target gene-specific mechanisms regulate Polycomb function during sequential cell-fate decisions.


Assuntos
Astrócitos/citologia , Diferenciação Celular , DNA Helicases/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Proteínas do Grupo Polycomb/metabolismo , Animais , Astrócitos/metabolismo , Linhagem Celular , Imunoprecipitação da Cromatina , DNA Helicases/genética , Embrião de Mamíferos , Proteína Potenciadora do Homólogo 2 de Zeste , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Proteína Glial Fibrilar Ácida , Histonas/química , Histonas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Complexo Repressor Polycomb 2/genética , Proteínas do Grupo Polycomb/genética , Gravidez , Regiões Promotoras Genéticas
19.
Cell Rep ; 2(6): 1554-62, 2012 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-23246003

RESUMO

The formation of the mammalian cortex requires the generation, migration, and differentiation of neurons. The vital role that the microtubule cytoskeleton plays in these cellular processes is reflected by the discovery that mutations in various tubulin isotypes cause different neurodevelopmental diseases, including lissencephaly (TUBA1A), polymicrogyria (TUBA1A, TUBB2B, TUBB3), and an ocular motility disorder (TUBB3). Here, we show that Tubb5 is expressed in neurogenic progenitors in the mouse and that its depletion in vivo perturbs the cell cycle of progenitors and alters the position of migrating neurons. We report the occurrence of three microcephalic patients with structural brain abnormalities harboring de novo mutations in TUBB5 (M299V, V353I, and E401K). These mutant proteins, which affect the chaperone-dependent assembly of tubulin heterodimers in different ways, disrupt neurogenic division and/or migration in vivo. Our results provide insight into the functional repertoire of the tubulin gene family, specifically implicating TUBB5 in embryonic neurogenesis and microcephaly.


Assuntos
Encéfalo/anormalidades , Encéfalo/metabolismo , Microcefalia/metabolismo , Mutação de Sentido Incorreto , Células-Tronco Neurais/metabolismo , Tubulina (Proteína)/metabolismo , Substituição de Aminoácidos , Animais , Encéfalo/embriologia , Encéfalo/patologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Mutantes , Microcefalia/embriologia , Microcefalia/genética , Microcefalia/patologia , Células-Tronco Neurais/patologia , Neurogênese/genética , Tubulina (Proteína)/genética
20.
Neuron ; 72(2): 269-84, 2011 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-22017987

RESUMO

Neurons in the mammalian neocortex arise from asymmetric divisions of progenitors residing in the ventricular zone. While in most progenitor divisions, the mitotic spindle is parallel to the ventricular surface, some progenitors reorient the spindle and divide in oblique orientations. Here, we use conditional deletion and overexpression of mouse Inscuteable (mInsc) to analyze the relevance of spindle reorientation in cortical progenitors. Mutating mInsc almost abolishes oblique and vertical mitotic spindles, while mInsc overexpression has the opposite effect. Our data suggest that oblique divisions are essential for generating the correct numbers of neurons in all cortical layers. Using clonal analysis, we demonstrate that spindle orientation affects the rate of indirect neurogenesis, a process where progenitors give rise to basal progenitors, which in turn divide symmetrically into two differentiating neurons. Our results indicate that the orientation of progenitor cell divisions is important for correct lineage specification in the developing mammalian brain.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Linhagem da Célula/fisiologia , Polaridade Celular/fisiologia , Neocórtex/crescimento & desenvolvimento , Neurogênese/fisiologia , Neurônios/metabolismo , Fuso Acromático/metabolismo , Alelos , Animais , Proteínas de Ciclo Celular/genética , Camundongos , Camundongos Transgênicos , Mutação , Neocórtex/metabolismo , Células-Tronco Neurais/metabolismo , Fuso Acromático/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...