Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Int J Mol Sci ; 24(8)2023 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-37108171

RESUMEN

Previous research has shown that cyclin-dependent kinases (Cdks) that play physiological roles in cell cycle regulation become activated in post-mitotic neurons after ischemic stroke, resulting in apoptotic neuronal death. In this article, we report our results using the widely used oxygen-glucose deprivation (OGD) in vitro model of ischemic stroke on primary mouse cortical neurons to investigate whether Cdk7, as part of the Cdk-activating kinase (CAK) complex that activates cell cycle Cdks, might be a regulator of ischemic neuronal death and may potentially constitute a therapeutic target for neuroprotection. We found no evidence of neuroprotection with either pharmacological or genetic invalidation of Cdk7. Despite the well-established idea that apoptosis contributes to cell death in the ischemic penumbra, we also found no evidence of apoptosis in the OGD model. This could explain the absence of neuroprotection following Cdk7 invalidation in this model. Neurons exposed to OGD seem predisposed to die in an NMDA receptor-dependent manner that could not be prevented further downstream. Given the direct exposure of neurons to anoxia or severe hypoxia, it is questionable how relevant OGD is for modeling the ischemic penumbra. Due to remaining uncertainties about cell death after OGD, caution is warranted when using this in vitro model to identify new stroke therapies.


Asunto(s)
Accidente Cerebrovascular Isquémico , Oxígeno , Ratones , Animales , Oxígeno/metabolismo , Glucosa/metabolismo , Apoptosis/genética , Muerte Celular/fisiología , Hipoxia , Quinasas Ciclina-Dependientes , Células Cultivadas
2.
Glia ; 70(9): 1652-1665, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35488490

RESUMEN

Mechanisms regulating oligodendrocyte differentiation, developmental myelination and myelin maintenance in adulthood are complex and still not completely described. Their understanding is crucial for the development of new protective or therapeutic strategies in demyelinating pathologies such as multiple sclerosis. In this perspective, we have investigated the role of Cyclin-dependent kinase 7 (Cdk7), a kinase involved in cell-cycle progression and transcription regulation, in the oligodendroglial lineage. We generated a conditional knock-out mouse model in which Cdk7 is invalidated in post-mitotic oligodendrocytes. At the end of developmental myelination, the number and diameter of myelinated axons, as well as the myelin structure, thickness and protein composition, were normal. However, in young adult and in aged mice, there was a higher number of small caliber myelinated axons associated with a decreased mean axonal diameter, myelin sheaths of large caliber axons were thinner, and the level of some major myelin-associated proteins was reduced. These defects were accompanied by the appearance of an abnormal clasping phenotype. We also used an in vitro oligodendroglial model and showed that Cdk7 pharmacological inhibition led to an altered myelination-associated morphological modification combined with a decreased expression of myelin-specific genes. Altogether, we identified novel functions for Cdk7 in CNS myelination.


Asunto(s)
Quinasas Ciclina-Dependientes , Vaina de Mielina , Oligodendroglía , Animales , Sistema Nervioso Central/metabolismo , Quinasas Ciclina-Dependientes/genética , Quinasas Ciclina-Dependientes/metabolismo , Expresión Génica , Ratones , Proteínas de la Mielina/metabolismo , Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo , Quinasa Activadora de Quinasas Ciclina-Dependientes
3.
Cell Mol Life Sci ; 77(22): 4553-4571, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32476056

RESUMEN

A persistent dogma in neuroscience supported the idea that terminally differentiated neurons permanently withdraw from the cell cycle. However, since the late 1990s, several studies have shown that cell cycle proteins are expressed in post-mitotic neurons under physiological conditions, indicating that the cell cycle machinery is not restricted to proliferating cells. Moreover, many studies have highlighted a clear link between cell cycle-related proteins and neurological disorders, particularly relating to apoptosis-induced neuronal death. Indeed, cell cycle-related proteins can be upregulated or overactivated in post-mitotic neurons in case of acute or degenerative central nervous system disease. Given the considerable lack of effective treatments for age-related neurological disorders, new therapeutic approaches targeting the cell cycle machinery might thus be considered. This review aims at summarizing current knowledge about the role of the cell cycle machinery in post-mitotic neurons in healthy and pathological conditions.


Asunto(s)
Ciclo Celular/fisiología , Mitosis/fisiología , Neuronas/fisiología , Animales , Apoptosis/fisiología , Proteínas de Ciclo Celular/metabolismo , Humanos , Enfermedades del Sistema Nervioso/metabolismo , Enfermedades del Sistema Nervioso/fisiopatología , Neuronas/metabolismo
4.
Cell Mol Life Sci ; 75(20): 3817-3827, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29728713

RESUMEN

Neural stem cells give rise to granule dentate neurons throughout life in the hippocampus. Upon activation, these stem cells generate fast proliferating progenitors that complete several rounds of divisions before differentiating into neurons. Although the mechanisms regulating the activation of stem cells have been intensively studied, little attention has been given so far to the intrinsic machinery allowing the expansion of the progenitor pool. The cell cycle protein Cdk6 positively regulates the proliferation of hippocampal progenitors, but the mechanism involved remains elusive. Whereas Cdk6 functions primarily as a cell cycle kinase, it can also act as transcriptional regulator in cancer cells and hematopoietic stem cells. Using mouse genetics, we show here that the function of Cdk6 in hippocampal neurogenesis relies specifically on its kinase activity. The present study also reveals a specific regulatory mechanism for Cdk6 in hippocampal progenitors. In contrast to the classical model of the cell cycle, we observe that the Cip/Kip family member p27, rather than the Ink4 family, negatively regulates Cdk6 in the adult hippocampus. Altogether, our data uncover a unique, cell type-specific regulatory mechanism controlling the expansion of hippocampal progenitors, where Cdk6 kinase activity is modulated by p27.


Asunto(s)
Proliferación Celular , Quinasa 6 Dependiente de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Animales , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/genética , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/deficiencia , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Giro Dentado/metabolismo , Giro Dentado/patología , Hipocampo/citología , Hipocampo/metabolismo , Hipocampo/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis
5.
Hippocampus ; 26(11): 1379-1392, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27325572

RESUMEN

In mammals, hippocampal dentate gyrus granule cells (DGCs) constitute a particular neuronal population produced both during embryogenesis and adult life, and play key roles in neural plasticity and memory. However, the molecular mechanisms regulating neurogenesis in the dentate lineage throughout development and adulthood are still not well understood. The Retinoblastoma protein (RB), a transcriptional repressor primarily involved in cell cycle control and cell death, plays crucial roles during cortical development but its function in the formation and maintenance of DGCs remains unknown. Here, we show that loss of RB during embryogenesis induces massive ectopic proliferation and delayed cell cycle exit of young DGCs specifically at late developmental stages but without affecting stem cells. This phenotype was partially counterbalanced by increased cell death. Similarly, during adulthood, loss of RB causes ectopic proliferation of newborn DGCs and dramatically impairs their survival. These results demonstrate a crucial role for RB in the generation and the survival of DGCs in the embryonic and the adult brain. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Giro Dentado/citología , Giro Dentado/embriología , Neurogénesis/genética , Neuronas/fisiología , Proteína de Retinoblastoma/metabolismo , Células Madre/fisiología , Animales , Diferenciación Celular/genética , Proliferación Celular/genética , Células Cultivadas , Factor de Transcripción E2F1/deficiencia , Factor de Transcripción E2F1/genética , Factor de Transcripción E2F3/genética , Factor de Transcripción E2F3/metabolismo , Embrión de Mamíferos , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Antígeno Ki-67/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Nestina/genética , Nestina/metabolismo , Proteína de Retinoblastoma/genética , Factores de Transcripción SOXB1/metabolismo
6.
Stem Cells ; 32(6): 1398-407, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24510844

RESUMEN

New cells are continuously generated from immature proliferating cells in the adult brain in two neurogenic niches known as the subgranular zone (SGZ) of the dentate gyrus (DG) of the hippocampus and the sub-ventricular zone (SVZ) of the lateral ventricles. However, the molecular mechanisms regulating their proliferation, differentiation, migration and functional integration of newborn neurons in pre-existing neural network remain largely unknown. Forkhead box (Fox) proteins belong to a large family of transcription factors implicated in a wide variety of biological processes. Recently, there has been accumulating evidence that several members of this family of proteins play important roles in adult neurogenesis. Here, we describe recent advances in our understanding of regulation provided by Fox factors in adult neurogenesis, and evaluate the potential role of Fox proteins as targets for therapeutic intervention in neurodegenerative diseases.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Neurogénesis , Transducción de Señal , Adulto , Animales , Secuencia de Bases , Factores de Transcripción Forkhead/química , Humanos , Datos de Secuencia Molecular
7.
J Neurosci ; 32(42): 14809-14, 2012 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-23077065

RESUMEN

The retinoblastoma protein (Rb) family members are essential regulators of cell cycle progression, principally through regulation of the E2f transcription factors. Growing evidence indicates that abnormal cell cycle signals can participate in neuronal death. In this regard, the role of Rb (p105) itself has been controversial. Germline Rb deletion leads to massive neuronal loss, but initial reports argue that death is non-cell autonomous. To more definitively resolve this question, we generated acute murine knock-out models of Rb in terminally differentiated neurons in vitro and in vivo. Surprisingly, we report that acute inactivation of Rb in postmitotic neurons results in ectopic cell cycle protein expression and neuronal loss without concurrent induction of classical E2f-mediated apoptotic genes, such as Apaf1 or Puma. These results suggest that terminally differentiated neurons require Rb for continuous cell cycle repression and survival.


Asunto(s)
Supervivencia Celular/fisiología , Mitosis/fisiología , Neuronas/fisiología , Proteína de Retinoblastoma/fisiología , Animales , Muerte Celular/genética , Muerte Celular/fisiología , Supervivencia Celular/genética , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitosis/genética , Proteína de Retinoblastoma/deficiencia
8.
Cell Mol Life Sci ; 69(9): 1493-503, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22068613

RESUMEN

The adult brain most probably reaches its highest degree of plasticity with the lifelong generation and integration of new neurons in the hippocampus and olfactory system. Neural precursor cells (NPCs) residing both in the subgranular zone of the dentate gyrus and in the subventricular zone of the lateral ventricles continuously generate neurons that populate the dentate gyrus and the olfactory bulb, respectively. The regulation of NPC proliferation in the adult brain has been widely investigated in the past few years. Yet, the intrinsic cell cycle machinery underlying NPC proliferation remains largely unexplored. In this review, we discuss the cell cycle components that are involved in the regulation of NPC proliferation in both neurogenic areas of the adult brain.


Asunto(s)
Ciclo Celular/fisiología , Neurogénesis/fisiología , Adulto , Animales , Encéfalo/citología , Encéfalo/crecimiento & desarrollo , Encéfalo/fisiología , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/fisiología , Ciclinas/fisiología , Humanos , Ratones , Modelos Neurológicos , Células-Madre Neurales/citología , Células-Madre Neurales/fisiología
9.
Stem Cells ; 29(4): 713-24, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21319271

RESUMEN

The presence of neurogenic precursors in the adult mammalian brain is now widely accepted, but the mechanisms coupling their proliferation with the onset of neuronal differentiation remain unknown. Here, we unravel the major contribution of the G(1) regulator cyclin-dependent kinase 6 (Cdk6) to adult neurogenesis. We found that Cdk6 was essential for cell proliferation within the dentate gyrus of the hippocampus and the subventricular zone of the lateral ventricles. Specifically, Cdk6 deficiency prevents the expansion of neuronally committed precursors by lengthening G(1) phase duration, reducing concomitantly the production of newborn neurons. Altogether, our data support G(1) length as an essential regulator of the switch between proliferation and neuronal differentiation in the adult brain and Cdk6 as one intrinsic key molecular regulator of this process.


Asunto(s)
Quinasa 6 Dependiente de la Ciclina/metabolismo , Fase G1 , Neurogénesis , Envejecimiento , Animales , Western Blotting , Diferenciación Celular , Proliferación Celular , Quinasa 4 Dependiente de la Ciclina/biosíntesis , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 6 Dependiente de la Ciclina/biosíntesis , Quinasa 6 Dependiente de la Ciclina/genética , Giro Dentado/citología , Giro Dentado/embriología , Técnica del Anticuerpo Fluorescente , Ventrículos Laterales/citología , Ventrículos Laterales/embriología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Neuronas/citología , Neuronas/fisiología
10.
Cell Rep ; 41(5): 111578, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36323247

RESUMEN

Long-term maintenance of the adult neurogenic niche depends on proper regulation of entry and exit from quiescence. Neural stem cell (NSC) transition from quiescence to activation is a complex process requiring precise cell-cycle control coordinated with transcriptional and morphological changes. How NSC fate transitions in coordination with the cell-cycle machinery remains poorly understood. Here we show that the Rb/E2F axis functions by linking the cell-cycle machinery to pivotal regulators of NSC fate. Deletion of Rb family proteins results in activation of NSCs, inducing a transcriptomic transition toward activation. Deletion of their target activator E2Fs1/3 results in intractable quiescence and cessation of neurogenesis. We show that the Rb/E2F axis mediates these fate transitions through regulation of factors essential for NSC function, including REST and ASCL1. Thus, the Rb/E2F axis is an important regulator of NSC fate, coordinating cell-cycle control with NSC activation and quiescence fate transitions.


Asunto(s)
Células Madre Adultas , Células-Madre Neurales , Células-Madre Neurales/metabolismo , Células Madre Adultas/metabolismo , Neurogénesis/fisiología , División Celular , Ciclo Celular , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo
11.
Gene Expr Patterns ; 9(5): 364-70, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19602392

RESUMEN

MicroRNAs (miRNAs) constitute a class of small non-coding endogenous RNAs that downregulate gene expression by binding to 3' untranslated region (UTR) of target messenger RNAs. Although they have been found to regulate developmental and physiological processes in several organs and tissues, their role in the regulation of the inner ear transcriptome remains unknown. In this report, we have performed systematic in situ hybridization to analyze the temporal and spatial distribution of three miRNAs (miR-96, mR-182, and mR-183) that are likely to arise from a single precursor RNA during the development and the maturation of the cochlea. Strikingly we found that the expression of mR-96, mR-182 and mR-183 was highly dynamic during the development of the cochlea, from the patterning to the differentiation of the main cochlear structures.


Asunto(s)
Oído Interno/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , MicroARNs/genética , Animales , Animales Recién Nacidos , Cóclea/embriología , Cóclea/crecimiento & desarrollo , Cóclea/metabolismo , Oído Interno/embriología , Oído Interno/crecimiento & desarrollo , Femenino , Técnica del Anticuerpo Fluorescente , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos BALB C , MicroARNs/metabolismo
12.
Curr Med Chem ; 16(6): 652-66, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19199929

RESUMEN

For a long time it was believed that the adult mammalian brain was completely unable to regenerate after insults. However, recent advances in the field of stem cell biology, including the identification of adult neural stem cells (NSCs) and evidence regarding a continuous production of neurons throughout life in the dentate gyrus (DG) and the subventricular zone of the lateral ventricles (SVZ), have provided new hopes for the development of novel therapeutic strategies to induce regeneration in the damaged brain. Moreover, proofs have accumulated this last decade that endogenous stem/progenitor cells of the adult brain have an intrinsic capacity to respond to brain disorders. Here, we first briefly summarize our current knowledge related to adult neurogenesis before focusing on the behaviour of adult neural stem/progenitors cells following stroke and seizure, and describe some of the molecular cues involved in the response of these cells to injury. In the second part, we outline the consequences of three main neurodegenerative disorders on adult neurogenesis and we discuss the potential therapeutic implication of adult neural stem/progenitors cells during the course of these diseases.


Asunto(s)
Enfermedades Neurodegenerativas/patología , Neurogénesis/fisiología , Neuronas/citología , Adulto , Animales , Humanos , Neuronas/patología
13.
BMC Neurosci ; 10: 30, 2009 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-19327139

RESUMEN

BACKGROUND: Newborn granule neurons are generated from proliferating neural stem/progenitor cells and integrated into mature synaptic networks in the adult dentate gyrus of the hippocampus. Since light/dark variations of the mitotic index and DNA synthesis occur in many tissues, we wanted to unravel the role of the clock-controlled Period2 gene (mPer2) in timing cell cycle kinetics and neurogenesis in the adult DG. RESULTS: In contrast to the suprachiasmatic nucleus, we observed a non-rhythmic constitutive expression of mPER2 in the dentate gyrus. We provide evidence that mPER2 is expressed in proliferating neural stem/progenitor cells (NPCs) and persists in early post-mitotic and mature newborn neurons from the adult DG. In vitro and in vivo analysis of a mouse line mutant in the mPer2 gene (Per2Brdm1), revealed a higher density of dividing NPCs together with an increased number of immature newborn neurons populating the DG. However, we showed that the lack of mPer2 does not change the total amount of mature adult-generated hippocampal neurons, because of a compensatory increase in neuronal cell death. CONCLUSION: Taken together, these data demonstrated a functional link between the constitutive expression of mPER2 and the intrinsic control of neural stem/progenitor cells proliferation, cell death and neurogenesis in the dentate gyrus of adult mice.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Proliferación Celular , Giro Dentado/citología , Hipocampo/citología , Neuronas/metabolismo , Proteínas Nucleares/fisiología , Células Madre/metabolismo , Factores de Transcripción/fisiología , Animales , Western Blotting , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Muerte Celular/fisiología , Células Cultivadas , Giro Dentado/crecimiento & desarrollo , Hipocampo/crecimiento & desarrollo , Inmunohistoquímica , Hibridación in Situ , Etiquetado Corte-Fin in Situ , Ratones , Ratones Noqueados , Neurogénesis/genética , Neurogénesis/fisiología , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Circadianas Period , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
14.
Cell Death Discov ; 4: 43, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29581894

RESUMEN

Cell cycle proteins are mainly expressed by dividing cells. However, it is well established that these molecules play additional non-canonical activities in several cell death contexts. Increasing evidence shows expression of cell cycle regulating proteins in post-mitotic cells, including mature neurons, following neuronal insult. Several cyclin-dependent kinases (Cdks) have already been shown to mediate ischemic neuronal death but Cdk1, a major cell cycle G2/M regulator, has not been investigated in this context. We therefore examined the role of Cdk1 in neuronal cell death following cerebral ischemia, using both in vitro and in vivo genetic and pharmacological approaches. Exposure of primary cortical neurons cultures to 4 h of oxygen-glucose deprivation (OGD) resulted in neuronal cell death and induced Cdk1 expression. Neurons from Cdk1-cKO mice showed partial resistance to OGD-induced neuronal cell death. Addition of R-roscovitine to the culture medium conferred neuroprotection against OGD-induced neuronal death. Transient 1-h occlusion of the cerebral artery (MCAO) also leads to Cdk1 expression and activation. Cdk1-cKO mice displayed partial resistance to transient 1-h MCAO. Moreover, systemic delivery of R-roscovitine was neuroprotective following transient 1-h MCAO. This study demonstrates that promising neuroprotective therapies can be considered through inhibition of the cell cycle machinery and particularly through pharmacological inhibition of Cdk1.

15.
Sci Rep ; 6: 20230, 2016 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-26847607

RESUMEN

Adult neural stem cells (aNSCs) are relatively quiescent populations that give rise to distinct neuronal subtypes throughout life, yet, at a very low rate and restricted differentiation potential. Thus, identifying the molecular mechanisms that control their cellular expansion is critical for regeneration after brain injury. Loss of the Retinoblastoma protein, Rb, leads to several defects in cell cycle as well as neuronal differentiation and migration during brain development. Here, we investigated the role of Rb during adult neurogenesis in the olfactory bulb (OB) by inducing its temporal deletion in aNSCs and progenitors. Loss of Rb was associated with increased proliferation of adult progenitors in the subventricular zone (SVZ) and the rostral migratory stream (RMS) but did not alter self-renewal of aNSCs or neuroblasts subsequent migration and terminal differentiation. Hence, one month after their birth, Rb-null neuroblasts were able to differentiate into distinct subtypes of GABAergic OB interneurons but were gradually lost after 3 months. Similarly, Rb controlled aNSCs/progenitors proliferation in vitro without affecting their differentiation capacity. This enhanced SVZ/OB neurogenesis associated with loss of Rb was only transient and negatively affected by increased apoptosis indicating a critical requirement for Rb in the long-term survival of adult-born OB interneurons.


Asunto(s)
Bulbo Olfatorio/citología , Proteína de Retinoblastoma/metabolismo , Animales , Antineoplásicos Hormonales/farmacología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular , Proteínas de Homeodominio/metabolismo , Hibridación Fluorescente in Situ , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis/efectos de los fármacos , Neuronas/metabolismo , Bulbo Olfatorio/metabolismo , Plásmidos/genética , Plásmidos/metabolismo , Proteína de Retinoblastoma/genética , Tamoxifeno/farmacología , Factores de Transcripción/metabolismo
16.
Front Neurosci ; 9: 458, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26696816

RESUMEN

Stroke affects one in every six people worldwide, and is the leading cause of adult disability. After stroke, some limited spontaneous recovery occurs, the mechanisms of which remain largely unknown. Multiple, parallel approaches are being investigated to develop neuroprotective, reparative and regenerative strategies for the treatment of stroke. For years, clinical studies have tried to use exogenous cell therapy as a means of brain repair, with varying success. Since the rediscovery of adult neurogenesis and the identification of adult neural stem cells in the late nineties, one promising field of investigation is focused upon triggering and stimulating this self-repair system to replace the neurons lost following brain injury. For instance, it is has been demonstrated that the adult brain has the capacity to produce large numbers of new neurons in response to stroke. The purpose of this review is to provide an updated overview of stroke-induced adult neurogenesis, from a cellular and molecular perspective, to its impact on brain repair and functional recovery.

17.
Cell Cycle ; 12(9): 1416-23, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23574720

RESUMEN

Neuronal survival is dependent upon the retinoblastoma family members, Rb1 (Rb) and Rb2 (p130). Rb is thought to regulate gene repression, in part, through direct recruitment of chromatin modifying enzymes to its conserved LXCXE binding domain. We sought to examine the mechanisms that Rb employs to mediate cell cycle gene repression in terminally differentiated cortical neurons. Here, we report that Rb loss converts chromatin at the promoters of E2f-target genes to an activated state. We established a mouse model system in which Rb-LXCXE interactions could be induciblely disabled. Surprisingly, this had no effect on survival or gene silencing in neuronal quiescence. Absence of the Rb LXCXE-binding domain in neurons is compatible with gene repression and long-term survival, unlike Rb deficiency. Finally, we are able to show that chromatin activation following Rb deletion occurs at the level of E2fs. Blocking E2f-mediated transcription downstream of Rb loss is sufficient to maintain chromatin in an inactive state. Taken together our results suggest a model whereby Rb-E2f interactions are sufficient to maintain gene repression irrespective of LXCXE-dependent chromatin remodeling.


Asunto(s)
Ciclo Celular , Ensamble y Desensamble de Cromatina , Factores de Transcripción E2F/metabolismo , Neuronas/citología , Neuronas/metabolismo , Proteína de Retinoblastoma/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Supervivencia Celular , Cromatina/metabolismo , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Regiones Promotoras Genéticas/genética , Unión Proteica , Estructura Terciaria de Proteína
18.
Front Cell Neurosci ; 7: 242, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24348336

RESUMEN

Neurotrophins are key players of neural development by controlling cell death programs. However, the signaling pathways that mediate their selective responses in different populations of neurons remain unclear. In the mammalian cochlea, sensory neurons differentiate perinatally into type I and II populations both expressing TrkB and TrkC, which bind respectively brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT3). How these two neuronal populations respond differentially to these two neurotrophins remains unknown. Here, we report in rat the segregation of the nuclear factor-κB (NFκB) subunit p65 specifically within the type II population postnatally. Using dissociated cultures of embryonic and postnatal spiral ganglion neurons, we observed a specific requirement of NFκB for BDNF but not NT3-dependent neuronal survival during a particular postnatal time window that corresponds to a period of neuronal cell death and hair cell innervation refinement in the developing cochlea. Consistently, postnatal p65 knockout mice showed a specific decreased number in type II spiral ganglion neurons. Taken together, these results identify NFκB as a type II neuron-specific factor that participates in the selective survival effects of BDNF and NT3 signaling on developing spiral ganglion neurons.

19.
Cell Stem Cell ; 12(4): 440-52, 2013 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-23499385

RESUMEN

The mechanisms through which cell-cycle control and cell-fate decisions are coordinated in proliferating stem cell populations are largely unknown. Here, we show that E2f3 isoforms, which control cell-cycle progression in cooperation with the retinoblastoma protein (pRb), have critical effects during developmental and adult neurogenesis. Loss of either E2f3 isoform disrupts Sox2 gene regulation and the balance between precursor maintenance and differentiation in the developing cortex. Both isoforms target the Sox2 locus to maintain baseline levels of Sox2 expression but antagonistically regulate Sox2 levels to instruct fate choices. E2f3-mediated regulation of Sox2 and precursor cell fate extends to the adult brain, where E2f3a loss results in defects in hippocampal neurogenesis and memory formation. Our results demonstrate a mechanism by which E2f3a and E2f3b differentially regulate Sox2 dosage in neural precursors, a finding that may have broad implications for the regulation of diverse stem cell populations.


Asunto(s)
Ciclo Celular , Factor de Transcripción E2F3/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Factores de Transcripción SOXB1/genética , Envejecimiento/metabolismo , Animales , Secuencia de Bases , Recuento de Células , Ciclo Celular/genética , Linaje de la Célula/genética , Proliferación Celular , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Ratones , Modelos Biológicos , Datos de Secuencia Molecular , Neurogénesis , Regiones Promotoras Genéticas/genética , Isoformas de Proteínas/metabolismo , Factores de Transcripción SOXB1/metabolismo
20.
J Cell Biol ; 193(2): 397-407, 2011 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-21502361

RESUMEN

The specific functions of intrinsic regulators of oligodendrocyte progenitor cell (OPC) division are poorly understood. Type 2 cyclin-dependent kinase (Cdk2) controls cell cycle progression of OPCs, but whether it acts during myelination and repair of demyelinating lesions remains unexplored. Here, we took advantage of a viable Cdk2(-/-) mutant mouse to investigate the function of this cell cycle regulator in OPC proliferation and differentiation in normal and pathological conditions. During central nervous system (CNS) development, Cdk2 loss does not affect OPC cell cycle, oligodendrocyte cell numbers, or myelination. However, in response to CNS demyelination, it clearly alters adult OPC renewal, cell cycle exit, and differentiation. Importantly, Cdk2 loss accelerates CNS remyelination of demyelinated axons. Thus, Cdk2 is dispensable for myelination but is important for adult OPC renewal, and could be one of the underlying mechanisms that drive adult progenitors to differentiate and thus regenerate myelin.


Asunto(s)
Sistema Nervioso Central/metabolismo , Quinasa 2 Dependiente de la Ciclina/genética , Neurogénesis/genética , Animales , Axones/metabolismo , Enfermedades Desmielinizantes/genética , Enfermedades Desmielinizantes/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Regeneración Nerviosa/fisiología , Células-Madre Neurales/metabolismo , Oligodendroglía/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA