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










Base de dados
Intervalo de ano de publicação
1.
EMBO Rep ; 25(5): 2418-2440, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38605277

RESUMO

Microcephaly is a common feature in inherited bone marrow failure syndromes, prompting investigations into shared pathways between neurogenesis and hematopoiesis. To understand this association, we studied the role of the microcephaly gene Mcph1 in hematological development. Our research revealed that Mcph1-knockout mice exhibited congenital macrocytic anemia due to impaired terminal erythroid differentiation during fetal development. Anemia's cause is a failure to complete cell division, evident from tetraploid erythroid progenitors with DNA content exceeding 4n. Gene expression profiling demonstrated activation of the p53 pathway in Mcph1-deficient erythroid precursors, leading to overexpression of Cdkn1a/p21, a major mediator of p53-dependent cell cycle arrest. Surprisingly, fetal brain analysis revealed hypertrophied binucleated neuroprogenitors overexpressing p21 in Mcph1-knockout mice, indicating a shared pathophysiological mechanism underlying both erythroid and neurological defects. However, inactivating p53 in Mcph1-/- mice failed to reverse anemia and microcephaly, suggesting that p53 activation in Mcph1-deficient cells resulted from their proliferation defect rather than causing it. These findings shed new light on Mcph1's function in fetal hematopoietic development, emphasizing the impact of disrupted cell division on neurogenesis and erythropoiesis - a common limiting pathway.


Assuntos
Proteínas de Ciclo Celular , Inibidor de Quinase Dependente de Ciclina p21 , Eritropoese , Camundongos Knockout , Microcefalia , Proteína Supressora de Tumor p53 , Animais , Eritropoese/genética , Microcefalia/genética , Microcefalia/patologia , Camundongos , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Mutação , Anemia Macrocítica/genética , Anemia Macrocítica/patologia , Anemia Macrocítica/metabolismo , Diferenciação Celular/genética , Células Precursoras Eritroides/metabolismo
2.
Neuron ; 109(3): 408-419, 2021 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-33306962

RESUMO

The neocortex, the seat of our higher cognitive abilities, has expanded in size during the evolution of certain mammals such as primates, including humans. This expansion occurs during development and is linked to the proliferative capacity of neural stem and progenitor cells (NPCs) in the neocortex. A number of cell-intrinsic and cell-extrinsic factors have been implicated in increasing NPC proliferative capacity. However, NPC metabolism has only recently emerged as major regulator of NPC proliferation. In this Perspective, we summarize recent insights into the role of NPC metabolism in neocortical development and neurodevelopmental disorders and its relevance for neocortex evolution. We discuss certain human-specific genes and microcephaly-implicated genes that operate in, or at, the mitochondria of NPCs and stimulate their proliferation by promoting glutaminolysis. We also discuss other metabolic pathways and develop a perspective on how metabolism mechanistically regulates NPC proliferation in neocortical development and how this contributed to neocortex evolution.


Assuntos
Evolução Biológica , Proliferação de Células/fisiologia , Redes e Vias Metabólicas/fisiologia , Neocórtex/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese/fisiologia , Animais , Humanos , Neurônios/fisiologia
3.
Cell Rep ; 31(2): 107506, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32294449

RESUMO

A distinctive feature of neocortical development is the highly coordinated production of different progenitor cell subtypes, which are critical for ensuring adequate neurogenic outcome and the development of normal neocortical size. To further understand the mechanisms that underlie neocortical growth, we focused our studies on the microcephaly gene Mcph1, and we report here that Mcph1 (1) exerts its functions in rapidly dividing apical radial glial cells (aRGCs) during mouse neocortical development stages that precede indirect neurogenesis; (2) is expressed at mitochondria; and (3) controls the proper proliferation and survival of RGCs, potentially through crosstalk with cellular metabolic pathways involving the stimulation of mitochondrial activity via VDAC1/GRP75 and AKT/HK2/VDAC1 and glutaminolysis via ATF4/PCK2. We currently report the description of a MCPH-gene implication in the interplay between bioenergetic pathways and neocortical growth, thus pointing to alterations of cellular metabolic pathways, in particular glutaminolysis, as a possible cause of microcephalic pathogenesis.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas do Citoesqueleto/genética , Microcefalia/genética , Microcefalia/metabolismo , Animais , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular/genética , Proliferação de Células/genética , Sobrevivência Celular/genética , Proteínas do Citoesqueleto/metabolismo , Feminino , Células HEK293 , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microcefalia/fisiopatologia , Mitocôndrias/metabolismo , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Mutação , Proteínas do Tecido Nervoso/metabolismo , Neurogênese/genética , Neuroglia/metabolismo , Neurônios/metabolismo , Canal de Ânion 1 Dependente de Voltagem/genética , Canal de Ânion 1 Dependente de Voltagem/metabolismo
4.
Cell Rep ; 29(3): 645-658.e5, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31618633

RESUMO

Changes in transcriptional regulation through cis-regulatory elements are thought to drive brain evolution. However, how this impacts the identity of primate cortical neurons is still unresolved. Here, we show that primate-specific cis-regulatory sequences upstream of the Dbx1 gene promote human-like expression in the mouse embryonic cerebral cortex, and this imparts cell identity. Indeed, while Dbx1 is expressed in highly restricted cortical progenitors in the mouse ventral pallium, it is maintained in neurons in primates. Phenocopy of the primate-like Dbx1 expression in mouse cortical progenitors induces ectopic Cajal-Retzius and subplate (SP) neurons, which are transient populations playing crucial roles in cortical development. A conditional expression solely in neurons uncouples mitotic and postmitotic activities of Dbx1 and exclusively promotes a SP-like fate. Our results highlight how transcriptional changes of a single fate determinant in postmitotic cells may contribute to the expansion of neuronal diversity during cortical evolution.


Assuntos
Evolução Biológica , Córtex Cerebral/metabolismo , Proteínas de Homeodomínio/metabolismo , Animais , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/patologia , Embrião de Mamíferos/metabolismo , Feminino , Proteínas de Homeodomínio/genética , Humanos , Macaca , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/metabolismo , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Gravidez , Proteínas com Domínio T/metabolismo
5.
Cereb Cortex ; 28(7): 2458-2478, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29722804

RESUMO

Neuropathological conditions might affect adult granulogenesis in the adult human dentate gyrus. However, radial glial cells (RGCs) have not been well characterized during human development and aging. We have previously described progenitor and neuronal layer establishment in the hippocampal pyramidal layer and dentate gyrus from embryonic life until mid-gestation. Here, we describe RGC subtypes in the hippocampus from 13 gestational weeks (GW) to mid-gestation and characterize their evolution and the dynamics of neurogenesis from mid-gestation to adulthood in normal and Alzheimer's disease (AD) subjects. In the pyramidal ventricular zone (VZ), RGC density declined with neurogenesis from mid-gestation until the perinatal period. In the dentate area, morphologic and antigenic differences among RGCs were observed from early ages of development to adulthood. Density and proliferative capacity of dentate RGCs as well as neurogenesis were strongly reduced during childhood until 5 years, few DCX+ cells are seen in adults. The dentate gyrus of both control and AD individuals showed Nestin+ and/or GFAPδ+ cells displaying different morphologies. In conclusion, pools of morphologically, antigenically, and topographically diverse neural progenitor cells are present in the human hippocampus from early developmental stages until adulthood, including in AD patients, while their neurogenic potential seems negligible in the adult.


Assuntos
Feto/citologia , Hipocampo , Células-Tronco Neurais/patologia , Neurogênese/fisiologia , Neurônios/patologia , Adolescente , Adulto , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer , Criança , Pré-Escolar , Feminino , Idade Gestacional , Hipocampo/embriologia , Hipocampo/crescimento & desenvolvimento , Hipocampo/patologia , Humanos , Lactente , Recém-Nascido , Antígeno Ki-67/metabolismo , Masculino , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Adulto Jovem
6.
Endocrinology ; 159(4): 1844-1859, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29509880

RESUMO

Pituitary growth hormone (GH) and insulinlike growth factor (IGF)-1 are anabolic hormones whose physiological roles are particularly important during development. The activity of the GH/IGF-1 axis is controlled by complex neuroendocrine systems including two hypothalamic neuropeptides, GH-releasing hormone (GHRH) and somatostatin (SRIF), and a gastrointestinal hormone, ghrelin. The neurotransmitter acetylcholine (ACh) is involved in tuning GH secretion, and its GH-stimulatory action has mainly been shown in adults but is not clearly documented during development. ACh, together with these hormones and their receptors, is expressed before birth, and somatotroph cells are already responsive to GHRH, SRIF, and ghrelin. We thus hypothesized that ACh could contribute to the modulation of the main components of the somatotropic axis during development. In this study, we generated a choline acetyltransferase knockout mouse line and showed that heterozygous mice display a transient deficit in ACh from embryonic day 18.5 to postnatal day 10, and they recover normal ACh levels from the second postnatal week. This developmental ACh deficiency had no major impact on weight gain and cardiorespiratory status of newborn mice. Using this mouse model, we found that endogenous ACh levels determined the concentrations of circulating GH and IGF-1 at embryonic and postnatal stages. In particular, serum GH level was correlated with brain ACh content. ACh also modulated the levels of GHRH and SRIF in the hypothalamus and ghrelin in the stomach, and it affected the levels of these hormones in the circulation. This study identifies ACh as a potential regulator of the somatotropic axis during the developmental period.


Assuntos
Acetilcolina/metabolismo , Colina O-Acetiltransferase/metabolismo , Hormônio do Crescimento/sangue , Hipotálamo/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Hipófise/metabolismo , Acetilcolina/sangue , Animais , Colina O-Acetiltransferase/genética , Mucosa Gástrica/metabolismo , Grelina/metabolismo , Hormônio Liberador de Hormônio do Crescimento/metabolismo , Heterozigoto , Camundongos , Camundongos Knockout , Sistemas Neurossecretores/metabolismo
7.
Cereb Cortex ; 27(1): 358-372, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-26443441

RESUMO

The molecular mechanisms that orchestrate the development of the human dentate gyrus are not known. In this study, we characterized the formation of human dentate and fimbrial progenitors and postmitotic neurons from 9 gestational weeks (GW9) to GW25. PAX6+ progenitor cells remained proliferative until GW16 in the dentate ventricular zone. By GW11, the secondary dentate matrix had developed in the intermediate zone, surrounding the dentate anlage and streaming toward the subpial layer. This secondary matrix contained proliferating PAX6+ and/or TBR2+ progenitors. In parallel, SOX2+ and PAX6+ fimbrial cells were detected approaching the dentate anlage, representing a possible source of extra-dentate progenitors. By GW16, when the granule cell layer could be delineated, a hilar matrix containing PAX6+ and some TBR2+ progenitors had become identifiable. By GW25, when the 2 limbs of the granule cell layer had formed, the secondary dentate matrix was reduced to a pool of progenitors at the fimbrio-dentate junction. Although human dentate development recapitulates key steps previously described in rodents, differences seemed to emerge in neuron layer markers expression. Further studies are necessary to better elucidate their role in dentate formation and connectivity.


Assuntos
Giro Denteado/embriologia , Fórnice/embriologia , Células-Tronco Neurais/citologia , Neurogênese/fisiologia , Neurônios/citologia , Biomarcadores/análise , Humanos
8.
Cereb Cortex ; 26(3): 1255-71, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25882041

RESUMO

The molecular mechanisms underlying the formation of hippocampus are unknown in humans. To improve our knowledge of molecules that potentially regulate pyramidal neurogenesis and layering in various hippocampal fields, we investigated the expression of progenitor markers and cell fate molecules from gestational week (GW) 9 to GW 20. At GW 9, the progenitor cell compartment of the hippocampal formation mainly consisted of PAX6(+) cells in the ventricular zone. Between GW 9 and 11, a second germinal area, the subventricular zone (SVZ), was formed, as shown by TBR2 labeling. Postmitotic markers (TBR1, CTIP2, SATB2, and CUX1) might reflect the inside-out layering of the plate from GW 11 onwards. TBR1(+) neurons appeared in the deep plate, whereas CTIP2(+), SATB2(+), and CUX1(+) neurons occupied the upper layers. From GW 16, differences in layer segregation were observed between the ammonic and subicular plates. Moreover, an ammonic-to-subicular maturation gradient was observed in germinal/postmitotic areas. Taken together, these findings demonstrate for the first time the presence of an SVZ in the hippocampus of human fetuses and laminar differences in transcription factor expression in the pyramidal layer of the human ammonic and subicular plate, and provide new information to further investigate the connectivity of the hippocampal formation.


Assuntos
Hipocampo/embriologia , Hipocampo/metabolismo , Células-Tronco Neurais/metabolismo , Células Piramidais/metabolismo , Proteínas do Olho/metabolismo , Hipocampo/citologia , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Microscopia Confocal , Microscopia de Fluorescência , Células-Tronco Neurais/citologia , Proteínas Nucleares/metabolismo , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/metabolismo , Células Piramidais/citologia , Proteínas Repressoras/metabolismo , Nicho de Células-Tronco/fisiologia , Proteínas com Domínio T/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo
9.
Neurobiol Dis ; 92(Pt A): 3-17, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26184894

RESUMO

Advances in the study of brain development over the last decades, especially recent findings regarding the evolutionary expansion of the human neocortex, and large-scale analyses of the proteome/transcriptome in the human brain, have offered novel insights into the molecular mechanisms guiding neural maturation, and the pathophysiology of multiple forms of neurological disorders. As a preamble to reviews of this issue, we provide an overview of the cellular, molecular and genetic bases of brain development with an emphasis on the major mechanisms associated with landmarks of normal neural development in the embryonic stage and early postnatal life, including neural stem/progenitor cell proliferation, cortical neuronal migration, evolution and folding of the cerebral cortex, synaptogenesis and neural circuit development, gliogenesis and myelination. We will only briefly depict developmental disorders that result from perturbations of these cellular or molecular mechanisms, and the most common perinatal brain injuries that could disturb normal brain development.


Assuntos
Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Animais , Encéfalo/citologia , Humanos , Transtornos do Neurodesenvolvimento/metabolismo
10.
Front Cell Neurosci ; 9: 92, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25870538

RESUMO

The development of the mammalian cerebral cortex involves a series of mechanisms: from patterning, progenitor cell proliferation and differentiation, to neuronal migration. Many factors influence the development of the cerebral cortex to its normal size and neuronal composition. Of these, the mechanisms that influence the proliferation and differentiation of neural progenitor cells are of particular interest, as they may have the greatest consequence on brain size, not only during development but also in evolution. In this context, causative genes of human autosomal recessive primary microcephaly, such as ASPM and MCPH1, are attractive candidates, as many of them show positive selection during primate evolution. MCPH1 causes microcephaly in mice and humans and is involved in a diverse array of molecular functions beyond brain development, including DNA repair and chromosome condensation. Positive selection of MCPH1 in the primate lineage has led to much insight and discussion of its role in brain size evolution. In this review, we will present an overview of MCPH1 from these multiple angles, and whilst its specific role in brain size regulation during development and evolution remain elusive, the pieces of the puzzle will be discussed with the aim of putting together the full picture of this fascinating gene.

11.
Dev Neurobiol ; 75(7): 721-37, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25369423

RESUMO

Motor activities are controlled by neural networks in the ventral spinal cord and consist in motor neurons and a set of distinct cardinal classes of spinal interneurons. These interneurons arise from distinct progenitor domains (p0-p3) delineated according to a transcriptional code. Neural progenitors of each domain express a unique combination of transcription factors (TFs) that largely contribute to determine the fate of four classes of interneurons (V0-V3) and motor neurons. In p2 domain, at least four subtypes of interneurons namely V2a, V2b, V2c, and Pax6(+) V2 are generated. Although genetic and molecular mechanisms that specify V2a and V2b are dependent on complex interplay between several TFs including Nkx6.1, Irx3, Gata2, Foxn4, and Ascl1, and signaling pathways such as Notch and TGF-ß, the sequence order of the activation of these regulators and their respective contribution are not completely elucidated yet. Here, we provide evidence by loss- or gain-of-function experiments that Gata2 is necessary for the normal development of both V2a and V2b neurons. We demonstrate that Nkx6.1 and Dll4 positively regulate the activation of Gata2 and Foxn4 in p2 progenitors. Gata2 also participates in the maintenance of p2 domain by repressing motor neuron differentiation and exerting a feedback control on patterning genes. Finally, Gata2 promotes the selective activation of V2b program at the expense of V2a fate. Thus our results provide new insights on the hierarchy and complex interactions between regulators of V2 genetic program.


Assuntos
Proteínas Aviárias/metabolismo , Fator de Transcrição GATA2/metabolismo , Interneurônios/fisiologia , Medula Espinal/embriologia , Medula Espinal/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Ligação ao Cálcio , Embrião de Galinha , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Fator de Transcrição GATA2/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Interneurônios/citologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Knockout , Camundongos Transgênicos , Neurônios Motores/citologia , Neurônios Motores/fisiologia , Neurogênese/fisiologia , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição Box Pareados/metabolismo , RNA não Traduzido/genética , RNA não Traduzido/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Medula Espinal/citologia , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/metabolismo
12.
Cell Rep ; 7(6): 1779-88, 2014 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-24931612

RESUMO

Over the course of cortical neurogenesis, the transition of progenitors from proliferation to differentiation requires a precise regulation of involved gene networks under varying environmental conditions. In order to identify such regulatory mechanisms, we analyzed microRNA (miRNA) target networks in progenitors during early and late stages of neurogenesis. We found that cyclin D1 is a network hub whose expression is miRNA-dosage sensitive. Experimental validation revealed a feedback regulation between cyclin D1 and its regulating miRNAs miR-20a, miR-20b, and miR-23a. Cyclin D1 induces expression of miR-20a and miR-20b, whereas it represses miR-23a. Inhibition of any of these miRNAs increases the developmental stage-specific mean and dynamic expression range (variance) of cyclin D1 protein in progenitors, leading to reduced neuronal differentiation. Thus, miRNAs establish robustness and stage-specific adaptability to a critical dosage-sensitive gene network during cortical neurogenesis. Understanding such network regulatory mechanisms for key developmental events can provide insights into individual susceptibilities for genetically complex neuropsychiatric disorders.


Assuntos
Redes Reguladoras de Genes , MicroRNAs/genética , Neurogênese/genética , Células-Tronco/citologia , Animais , Diferenciação Celular/genética , Humanos , Camundongos , Camundongos Transgênicos
13.
J Neuropathol Exp Neurol ; 73(2): 143-58, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24423639

RESUMO

Neurologic morbidity associated with congenital cytomegalovirus (CMV) infection is a major public health concern. The pathogenesis of cerebral lesions remains unclear. We report the neuropathologic substrates, the immune response, and the cellular targets of CMV in 16 infected human fetal brains aged 23 to 28.5 gestational weeks. Nine cases were microcephalic, 10 had extensive cortical lesions, 8 had hippocampal abnormalities, and 5 cases showed infection of the olfactory bulb. The density of CMV-immunolabeled cells correlated with the presence of microcephaly and the extent of brain abnormalities. Innate and adaptive immune responses were present but did not react against all CMV-infected cells. Cytomegalovirus infected all cell types but showed higher tropism for stem cells/radial glial cells. The results indicate that 2 main factors influence the neuropathologic outcome at this stage: the density of CMV-positive cells and the tropism of CMV for stem/progenitor cells. This suggests that the large spectrum of CMV-induced brain abnormalities is caused not only by tissue destruction but also by the particular vulnerability of stem cells during early brain development. Florid infection of the hippocampus and the olfactory bulb may expose these patients to the risk of neurocognitive and sensorineural handicap even in cases of infection at late stages of gestation.


Assuntos
Encéfalo/embriologia , Encéfalo/patologia , Infecções por Citomegalovirus/patologia , Citomegalovirus/patogenicidade , Encéfalo/metabolismo , Encéfalo/virologia , Estudos de Casos e Controles , Feto , Idade Gestacional , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Antígeno Ki-67/metabolismo
14.
Acta Neuropathol ; 122(6): 763-74, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22033878

RESUMO

Congenital cytomegalovirus (CMV) infection is the leading cause of non-hereditary congenital sensorineural hearing loss (SNHL). The natural course and the pathophysiology of inner ear lesions during human fetal CMV infection have not yet been reported. Inner ear lesions were investigated in six CMV-infected fetuses aged 19-35 postconceptional weeks and correlated with central nervous system (CNS) lesions. All the fetuses had high viral loads in the amniotic fluid and severe visceral and CNS lesions visible by ultrasound. Diffuse lesions consisting of both cytomegalic cells containing inclusion bodies and inflammation were found within all studied structures including the inner ear, brain, other organs, and placenta, suggesting hematogenous dissemination. Cochlear infection was consistently present and predominated in the stria vascularis (5/6), whereas the supporting cells in the organ of Corti were less often involved (2/6). Vestibular infection, found in 4/6 cases, was florid; the non-sensory epithelia, including the dark cells, were extensively infected. The endolymphatic sac was infected in 1 of 3 cases. The severity of inner ear infection was correlated with the CNS lesions, confirming the neurotropism of CMV. This study documenting infection of the structures involved in endolymph secretion and potassium homeostasis in fetuses with high amniotic fluid viral loads suggests that potassium dysregulation in the endolymphatic compartment of the inner ear may lead to secondary degeneration of the sensory structures. In addition, the occurrence of SNHL depends on the intensity and duration of the viral infection and inflammation.


Assuntos
Infecções por Citomegalovirus/congênito , Infecções por Citomegalovirus/patologia , Doenças Fetais/patologia , Doenças Fetais/virologia , Feto/virologia , Doenças do Labirinto/congênito , Doenças do Labirinto/virologia , Líquido Amniótico/virologia , Autopsia , Estudos de Casos e Controles , Doenças do Sistema Nervoso Central/congênito , Doenças do Sistema Nervoso Central/patologia , Doenças do Sistema Nervoso Central/virologia , Cóclea/patologia , Cóclea/virologia , Infecções por Citomegalovirus/metabolismo , Saco Endolinfático/patologia , Saco Endolinfático/virologia , Feminino , Doenças Fetais/metabolismo , Homeostase , Humanos , Doenças do Labirinto/patologia , Órgão Espiral/patologia , Órgão Espiral/virologia , Potássio/metabolismo , Gravidez , Segundo Trimestre da Gravidez , Terceiro Trimestre da Gravidez , Estudos Retrospectivos , Vestíbulo do Labirinto/patologia , Vestíbulo do Labirinto/virologia , Carga Viral
15.
J Clin Invest ; 121(8): 3071-87, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21737879

RESUMO

Autosomal recessive primary microcephaly (MCPH) is a genetic disorder that causes a reduction of cortical outgrowth without severe interference with cortical patterning. It is associated with mutations in a number of genes encoding protein involved in mitotic spindle formation and centrosomal activities or cell cycle control. We have shown previously that blocking vasoactive intestinal peptide (VIP) during gestation in mice by using a VIP antagonist (VA) results in microcephaly. Here, we have shown that the cortical abnormalities caused by prenatal VA administration mimic the phenotype described in MCPH patients and that VIP blockade during neurogenesis specifically disrupts Mcph1 signaling. VA administration reduced neuroepithelial progenitor proliferation by increasing cell cycle length and promoting cell cycle exit and premature neuronal differentiation. Quantitative RT-PCR and Western blot showed that VA downregulated Mcph1. Inhibition of Mcph1 expression led to downregulation of Chk1 and reduction of Chk1 kinase activity. The inhibition of Mcph1 and Chk1 affected the expression of a specific subset of cell cycle­controlling genes and turned off neural stem cell proliferation in neurospheres. Furthermore, in vitro silencing of either Mcph1 or Chk1 in neurospheres mimicked VA-induced inhibition of cell proliferation. These results demonstrate that VIP blockade induces microcephaly through Mcph1 signaling and suggest that VIP/Mcph1/Chk1 signaling is key for normal cortical development.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Regulação da Expressão Gênica , Microcefalia/metabolismo , Proteínas Quinases/metabolismo , Transdução de Sinais , Peptídeo Intestinal Vasoativo/fisiologia , Animais , Ciclo Celular , Proteínas de Ciclo Celular , Diferenciação Celular , Proliferação de Células , Quinase 1 do Ponto de Checagem , Proteínas do Citoesqueleto , Feminino , Camundongos , Modelos Biológicos , Neurônios/metabolismo , Células-Tronco/citologia , Peptídeo Intestinal Vasoativo/metabolismo
16.
Stem Cells ; 27(7): 1643-53, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19544452

RESUMO

The identification of the factors that allow better monitoring of stem cell renewal and differentiation is of paramount importance for the implementation of new regenerative therapies, especially with regard to the nervous and hematopoietic systems. In this article, we present new information on the function of zinc finger protein 191 (ZNF/Zfp191), a factor isolated in hematopoietic cell lines, within progenitors of the central nervous system (CNS). ZNF/Zfp191 has been found to be principally expressed in progenitors of the developing CNS of humans and mice. Such an overlap of the expression patterns in addition to the high homology of the protein in mammals suggested that ZNF/Zfp191 exerts a conserved function within such progenitors. Indeed, ZNF191 knockdown in human neural progenitors inhibits proliferation and leads to the exit of the cell cycle. Conversely, ZNF191 misexpression maintains progenitors in cycle and exerts negative control on the Notch pathway, which prevents them from differentiating. The present data, together with the fact that the inactivation of Zfp191 leads to embryonic lethality, confirm ZNF191 as an essential factor acting for the promotion of the cell cycle and thus maintenance in the progenitor stage. On the bases of expression data, such a function can be extended to progenitor cells of other tissues such as the hematopoietic system, which emphasizes the important issue of further understanding the molecular events controlled by ZNF/Zfp191.


Assuntos
Proteínas de Transporte/fisiologia , Fatores de Transcrição Kruppel-Like/fisiologia , Neurônios/citologia , Neurônios/metabolismo , Células-Tronco/citologia , Animais , Proteínas de Transporte/genética , Ciclo Celular/genética , Ciclo Celular/fisiologia , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Sistema Nervoso Central/embriologia , Sistema Nervoso Central/metabolismo , Embrião de Galinha , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Feminino , Citometria de Fluxo , Humanos , Hibridização In Situ , Fatores de Transcrição Kruppel-Like/genética , Lentivirus/genética , Masculino , Camundongos , Gravidez , Receptores Notch/genética , Receptores Notch/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Células-Tronco/metabolismo
17.
Gene Expr Patterns ; 8(3): 148-54, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18096443

RESUMO

The human zinc finger protein 191 (ZNF191) is a Krüppel-like protein and can specifically interact with the widespread TCAT motif which constitutes the HUMTH01 microsatellite in the tyrosine hydroxylase (TH) gene (encoding the rate-limiting enzyme in the synthesis of catecholamines). Allelic variations of HUMTH01 are known to have a quantitative silencing effect on TH gene expression and to correlate with quantitative and qualitative changes in the binding by ZNF191. This factor has been isolated from bone marrow and promyelocytic leukemia cell lines indicating that ZNF191 also plays a role in hematopoiesis. Thus, ZNF191 could participate in the regulation of several genes implicated in different functions. Moreover, mice that are deficient in Zfp191, the murine homologue of ZNF191, have been shown to be severely retarded in development and to die approximately at embryonic day 7.5. In order to gain further insight into its biological functions, we have analysed the localisation of Zfp191 throughout mouse development. Expression was detected early during embryogenesis in ectodermal, endodermal, mesodermal and extra-embryonic tissues. In particular, Zfp191 was observed in the developing central nervous system. Interestingly, its expression levels were prominent in areas of proliferation such as the subventricular zone. Zfp191 expression pattern during development can account for the phenotypic features of Zfp191(-/-) embryos.


Assuntos
Proteínas de Transporte/biossíntese , Proteínas de Transporte/genética , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Animais , Desenvolvimento Embrionário/fisiologia , Feminino , Camundongos , Sistema Nervoso/embriologia , Sistema Nervoso/metabolismo , Fenótipo , Dedos de Zinco/genética
18.
Development ; 133(11): 2155-65, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16672344

RESUMO

Postmitotic neurons are produced from a pool of cycling progenitors in an orderly fashion that requires proper spatial and temporal coordination of proliferation, fate determination, differentiation and morphogenesis. This probably relies on complex interplay between mechanisms that control cell cycle, specification and differentiation. In this respect, we have studied the possible implication of GATA2, a transcription factor that is involved in several neuronal specification pathways, in the control of the proliferation of neural progenitors in the embryonic spinal cord. Using gain- and loss-of-function manipulations, we have shown that Gata2 can drive neural progenitors out of the cycle and, to some extent, into differentiation. This correlates with the control of cyclin D1 transcription and of the expression of the p27/Kip1 protein. Interestingly, this functional aspect is not only associated with silencing of the Notch pathway but also appears to be independent of proneural function. Consistently, GATA2 also controls the proliferation capacity of mouse embryonic neuroepithelial cells in culture. Indeed, Gata2 inactivation enhances the proliferation rate in these cells. By contrast, GATA2 overexpression is sufficient to force such cells and neuroblastoma cells to stop dividing but not to drive either type of cell into differentiation. Furthermore, a non-cell autonomous effect of Gata2 expression was observed in vivo as well as in vitro. Hence, our data have provided evidence for the ability of Gata2 to inhibit the proliferation of neural progenitors, and they further suggest that, in this regard, Gata2 can operate independently of neuronal differentiation.


Assuntos
Fator de Transcrição GATA2/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Animais Recém-Nascidos , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Embrião de Galinha , Ciclina D1/metabolismo , Ciclina D2 , Ciclinas/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Fator de Transcrição GATA2/deficiência , Fator de Transcrição GATA2/genética , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Notch/metabolismo , Transdução de Sinais
19.
J Neurosci Res ; 73(6): 737-52, 2003 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-12949900

RESUMO

Neuroepithelial b2T cells were derived from the hindbrain and the spinal cord of mouse transgenic embryos, which expressed SV40 T antigen under the control of a Hoxb2 enhancer. Strikingly, b2T cell lines of either origin exhibit a very similar gene expression pattern, including markers of the hindbrain and the spinal cord, such as Hox genes, but not of more anterior cephalic regions. In addition, the broad expression pattern of b2T cells, probably linked to culture conditions, appeared to be appropriately modulated when the cells were reimplanted at different longitudinal levels into chick host embryos, suggesting that these cells are responsive to exogenous signalling mechanisms. Further support for these allegations was obtained by culturing b2T cells in defined medium and by assessing the expression of Krox20, an odd-numbered rhombomere marker, which appeared to be modulated by a complex interplay between FGF, retinoic acid (RA), and noggin. With respect to these as yet unique properties, b2T cells constitute an original alternative tool to in vivo models for the analysis of molecular pathways involved in the patterning of the neural tube.


Assuntos
Proteínas Aviárias , Padronização Corporal/fisiologia , Linhagem Celular , Proteínas do Tecido Nervoso , Rombencéfalo/citologia , Medula Espinal/citologia , Animais , Antígenos Transformantes de Poliomavirus/metabolismo , Antineoplásicos/farmacologia , Proteínas de Transporte , Células Cultivadas , Embrião de Galinha , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Relação Dose-Resposta a Droga , Interações Medicamentosas , Proteína 2 de Resposta de Crescimento Precoce , Embrião de Mamíferos , Elementos Facilitadores Genéticos/fisiologia , Epitélio/metabolismo , Fatores de Crescimento de Fibroblastos/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Proteínas de Filamentos Intermediários/metabolismo , Fator de Transcrição MafB , Mesencéfalo/metabolismo , Camundongos , Camundongos Transgênicos , Morfogênese , Nestina , Crista Neural , Proteínas Oncogênicas/metabolismo , Prosencéfalo/metabolismo , Proteínas/farmacologia , Sondas RNA/metabolismo , Rombencéfalo/embriologia , Rombencéfalo/metabolismo , Medula Espinal/embriologia , Medula Espinal/metabolismo , Transativadores/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Tretinoína/farmacologia
20.
EMBO J ; 21(3): 365-76, 2002 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11823429

RESUMO

In the segmented vertebrate hindbrain, the Hoxa3 and Hoxb3 genes are expressed at high relative levels in the rhombomeres (r) 5 and 6, and 5, respectively. The single enhancer elements responsible for these activities have been identified previously and shown to constitute direct targets of the transcription factor kreisler, which is expressed in r5 and r6. Here, we have analysed the contribution of the transcription factor Krox20, present in r3 and r5. Genetic analyses demonstrated that Krox20 is required for activity of the Hoxb3 r5 enhancer, but not of the Hoxa3 r5/6 enhancer. Mutational analysis of the Hoxb3 r5 enhancer, together with ectopic expression experiments, revealed that Krox20 binds to the enhancer and synergizes with kreisler to promote Hoxb3 transcription, restricting enhancer activity to their domain of overlap, r5. These analyses also suggested contributions from an Ets-related factor and from putative factors likely to heterodimerize with kreisler. The integration of multiple independent inputs present in overlapping domains by a single enhancer is likely to constitute a general mechanism for the patterning of subterritories during vertebrate development.


Assuntos
Proteínas Aviárias , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Proteínas Oncogênicas , Rombencéfalo/fisiologia , Fatores de Transcrição/genética , Proteínas de Xenopus , Animais , Análise Mutacional de DNA , Proteína 2 de Resposta de Crescimento Precoce , Fator de Transcrição MafB , Camundongos , Rombencéfalo/embriologia , Ativação Transcricional , Dedos de Zinco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...