Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
EMBO J ; 43(3): 317-338, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38177500

RESUMEN

Lifelong hippocampal neurogenesis is maintained by a pool of multipotent adult neural stem cells (aNSCs) residing in the subgranular zone of the dentate gyrus (DG). The mechanisms guiding transition of NSCs from the developmental to the adult state remain unclear. We show here, by using nestin-based reporter mice deficient for cyclin D2, that the aNSC pool is established through cyclin D2-dependent proliferation during the first two weeks of life. The absence of cyclin D2 does not affect normal development of the dentate gyrus until birth but prevents postnatal formation of radial glia-like aNSCs. Furthermore, retroviral fate mapping reveals that aNSCs are born on-site from precursors located in the dentate gyrus shortly after birth. Taken together, our data identify the critical time window and the spatial location of the precursor divisions that generate the persistent population of aNSCs and demonstrate the central role of cyclin D2 in this process.


Asunto(s)
Células-Madre Neurales , Neuronas , Animales , Ratones , Ciclina D2/genética , Giro Dentado , Hipocampo , Neurogénesis
2.
Brain ; 146(8): 3416-3430, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-36825472

RESUMEN

Epilepsy, characterized by recurrent unprovoked seizures resulting from a wide variety of causes, is one of the world's most prominent neurological disabilities. Seizures, which are an expression of neuronal network dysfunction, occur in a positive feedback loop of concomitant factors, including neuro-inflammatory responses, where seizures generate more seizures. Among other pathways involved in inflammatory responses, the JAK/STAT signalling pathway has been proposed to participate in epilepsy. Here, we tested an in vitro model of temporal lobe epilepsy, with the hypothesis that acute blockage of STAT3-phosphorylation during epileptogenesis would prevent structural damage in the hippocampal circuitry and the imprinting of both neural epileptic activity and inflammatory glial states. We performed calcium imaging of spontaneous circuit dynamics in organotypic hippocampal slices previously exposed to epileptogenic conditions through the blockage of GABAergic synaptic transmission. Epileptogenic conditions lead to epileptic dynamics imprinted on circuits in terms of increased neuronal firing and circuit synchronization, increased correlated activity in neuronal pairs and decreased complexity in synchronization patterns. Acute blockage of the STAT3-phosphorylation during epileptogenesis prevented the imprinting of epileptic activity patterns, general cell loss, loss of GABAergic neurons and the persistence of reactive glial states. This work provides mechanistic evidence that blocking the STAT3 signalling pathway during epileptogenesis can prevent patho-topological persistent reorganization of neuro-glial circuits.


Asunto(s)
Epilepsia del Lóbulo Temporal , Epilepsia , Humanos , Convulsiones/metabolismo , Epilepsia del Lóbulo Temporal/metabolismo , Hipocampo/metabolismo , Neuronas GABAérgicas/metabolismo , Factor de Transcripción STAT3/metabolismo
3.
Front Neurosci ; 17: 1186256, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37496737

RESUMEN

Hippocampal neurogenesis is a tightly regulated process in which neural stem cells (NSCs) get activated, enter in the cell cycle and give rise to neurons after a multistep process. Quiescent and activated NSCs, neural precursors, immature and mature neurons and newborn astrocytes coexist in the neurogenic niche in a strictly controlled environment which maintains the correct functioning of neurogenesis. NSCs are the first step in the neurogenic process and are a finite and, mostly, non-renewable resource, therefore any alteration of the intrinsic properties of NSCs will impact the total neurogenic output. Neuronal hyperexcitation is a strong activator of NSCs prompting them to divide and therefore increasing neurogenesis. However, neuronal hyperactivity is not an isolated process but often also involves excitotoxicity which is subsequently accompanied by neuroinflammation. Neuroinflammation normally reduces the activation of NSCs. It is technically difficult to isolate the effect of neuronal hyperexcitation alone, but neuroinflammation without neuronal hyperexcitation can be studied in a variety of models. In order to shed light on how the balance of neuronal hyperexcitation and neuroinflammation affect NSCs we analyzed proliferation and morphology of NSCs. We used two models of neuronal hyperactivity [an epilepsy model induced by KA, and a model of traumatic brain injury (TBI)] and different models of inflammation (LPS, Poly I:C, IFN-α and IL-6). We observed that only those models that induce neuronal hyperactivity induce NSCs activation but neuroinflammation causes the opposite effect. We also analyzed the response of other cell types in the neurogenic niche, focusing on astrocytes.

4.
Cell Stem Cell ; 28(5): 783-785, 2021 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-33961757

RESUMEN

Hippocampal neural stem cells (NSCs) and neurogenesis decline sharply with age, though a small population remains. Two articles in this issue of Cell Stem Cell by Ibrayeva et al. (2021) and Harris et al. (2021) indicate the presence of subpopulations of NSCs whose dynamics of activation and self-renewal change over time and may be key to NSC preservation.


Asunto(s)
Células Madre Adultas , Células-Madre Neurales , Proliferación Celular , Hipocampo , Neurogénesis
5.
Front Cell Dev Biol ; 8: 654, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32793597

RESUMEN

Hippocampal neurogenesis, the process by which neural stem cells (NSCs) continuously generate new neurons in the dentate gyrus (DG) of most mammals including humans, is chiefly regulated by neuronal activity. Thus, severe alterations have been found in samples from epilepsy patients and in the hippocampal neurogenic niche in mouse models of epilepsy. Reactive-like and gliogenic NSCs plus aberrant newborn neurons with altered migration, morphology, and functional properties are induced by seizures in experimental models of temporal lobe epilepsy. Hippocampal neurogenesis participates in memory and learning and in the control of anxiety and stress. It has been therefore hypothesized that part of the cognitive symptoms associated with epilepsy could be promoted by impaired hippocampal neurogenesis. We here analyze for the first time the alterations of the neurogenic niche in a novel mouse model of Dravet syndrome (DS), a genetic encephalopathy with severe epilepsy in infancy and multiple neurological comorbidities. Scn1aWT/A1783V mice, hereafter referred to as DS, carrying a heterozygous and clinically relevant SCN1A mutation (A1783V) recapitulate the disease at the genetic and phenotypic levels. We demonstrate that in the neurogenic niche of young adult DS mice there are fewer NSCs, they have impaired cell division and bear reactive-like morphology. In addition, there is significant aberrant neurogenesis. Newborn immature neurons migrate abnormally, and several morphological features are drastically changed. Thus, this study shows for the first time important modifications in hippocampal neurogenesis in DS and opens venues for further research on this topic.

6.
Front Mol Neurosci ; 13: 602801, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33584198

RESUMEN

Dravet syndrome (DS) is an epileptic syndrome caused by mutations in the Scn1a gene encoding the α1 subunit of the sodium channel Nav1.1, which is associated with febrile seizures that progress to severe tonic-clonic seizures and associated comorbidities. Treatment with cannabidiol has been approved to reduce seizures in DS, but it may also be active against these comorbidities. The aim of this study was to validate a new mouse model of DS having lower mortality than previous models, which may serve to further evaluate therapies for the long-term comorbidities. This new model consists of heterozygous conditional knock-in mice carrying a missense mutation (A1783V) in Scn1a gene expressed exclusively in neurons of the CNS (Syn-Cre/Scn1aWT/A1783V). These mice have been used here to determine the extent and persistence of the behavioral deterioration in different postnatal days (PND), as well as to investigate the alterations that the disease produces in the endocannabinoid system and the contribution of inflammatory events and impaired neurogenesis in the pathology. Syn-Cre/Scn1aWT/A1783V mice showed a strong reduction in hindlimb grasp reflex at PND10, whereas at PND25, they presented spontaneous convulsions and a greater susceptibility to pentylenetetrazole-induced seizures, marked hyperactivity, deficient spatial working memory, lower levels of anxiety, and altered social interaction behavior. These differences disappeared at PND40 and PND60, except the changes in social interaction and anxiety. The analysis of CNS structures associated with these behavioral alterations revealed an elevated glial reactivity in the prefrontal cortex and the dentate gyrus. This was associated in the dentate gyrus with a greater cell proliferation detected with Ki67 immunostaining, whereas double-labeling analyses identified that proliferating cells were GFAP-positive suggesting failed neurogenesis but astrocyte proliferation. The analysis of the endocannabinoid system of Syn-Cre/Scn1aWT/A1783V mice confirmed reductions in CB1 receptors and MAGL and FAAH enzymes, mainly in the cerebellum but also in other areas, whereas CB2 receptors became upregulated in the hippocampus. In conclusion, Syn-Cre/Scn1aWT/A1783V mice showed seizuring susceptibility and several comorbidities (hyperactivity, memory impairment, less anxiety, and altered social behavior), which exhibited a pattern of age expression similar to DS patients. Syn-Cre/Scn1aWT/A1783V mice also exhibited greater glial reactivity and a reactive response in the neurogenic niche, and regional changes in the status of the endocannabinoid signaling, events that could contribute in behavioral impairment.

7.
Front Neurosci ; 14: 811, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32922255

RESUMEN

A population of neural stem cells (NSCs) dwelling in the dentate gyrus (DG) is able to generate neurons throughout adult life in the hippocampus of most mammals. These NSCs generate also astrocytes naturally and are capable of generating oligodendrocytes after gene manipulation. It has been more recently shown that adult hippocampal NSCs after epileptic seizures as well as subventricular zone NSCs after stroke can give rise to reactive astrocytes (RAs). In the hippocampus, the induction of seizures triggers the conversion of NSCs into reactive NSCs (React-NSCs) characterized by a drastic morphological transformation, abnormal migration, and massive activation or entry into the cell cycle to generate more React-NSCs that ultimately differentiate into RAs. In the search for tools to investigate the properties of React-NSCs, we have explored the LPA1-green fluorescent protein (GFP) transgenic line of mice in which hippocampal NSCs are specifically labeled due to the expression of lysophosphatidic acid receptor 1 (LPA1). We first addressed the validity of the transgene expression as true marker of LPA1 expression and then demonstrated how, after seizures, LPA1-GFP labeled exclusively React-NSCs for several weeks. Then React-NSCs lost LPA1-GFP expression as neurons of the granule cell layer started to express it. Finally, we used knockout for LPA1 transgenic mice to show that LPA1 plays a functional role in the activation of React-NSCs. Thus, we confirmed that LPA1-GFP expression is a valid tool to study both NSCs and React-NSCs and that the LPA1 pathway could be a target in the intent to preserve NSCs after seizures.

9.
Aging Cell ; 18(4): e12958, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30989815

RESUMEN

Adult neurogenesis persists in the hippocampus of most mammal species during postnatal and adult life, including humans, although it declines markedly with age. The mechanisms driving the age-dependent decline of hippocampal neurogenesis are yet not fully understood. The progressive loss of neural stem cells (NSCs) is a main factor, but the true neurogenic output depends initially on the actual number of activated NSCs in each given time point. Because the fraction of activated NSCs remains constant relative to the total population, the real number of activated NSCs declines in parallel to the total NSC pool. We investigated aging-associated changes in NSCs and found that there are at least two distinct populations of NSCs. An alpha type, which maintains the classic type-1 radial morphology and accounts for most of the overall NSC mitotic activity; and an omega type characterized by increased reactive-like morphological complexity and much lower probability of division even under a pro-activation challenge. Finally, our results suggest that alpha-type NSCs are able to transform into omega-type cells overtime and that this phenotypic and functional change might be facilitated by the chronic inflammation associated with aging.


Asunto(s)
Senescencia Celular/fisiología , Giro Dentado/citología , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Fenotipo , Animales , Antiinflamatorios/farmacología , Diferenciación Celular/fisiología , Inflamación/metabolismo , Ratones , Ratones Transgénicos , Minociclina/farmacología , Mitosis/fisiología , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/fisiología
10.
Front Cell Dev Biol ; 7: 158, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31482091

RESUMEN

Adult neurogenesis persists in the adult hippocampus due to the presence of multipotent neural stem cells (NSCs). Hippocampal neurogenesis is involved in a range of cognitive functions and is tightly regulated by neuronal activity. NSCs respond promptly to physiological and pathological stimuli altering their neurogenic and gliogenic potential. In a mouse model of mesial temporal lobe epilepsy (MTLE), seizures triggered by the intrahippocampal injection of the glutamate receptor agonist kainic acid (KA) induce NSCs to convert into reactive NSCs (React-NSCs) which stop producing new neurons and ultimately generate reactive astrocytes thus contributing to the development of hippocampal sclerosis and abolishing neurogenesis. We herein show how seizures triggered by the injection of KA in the amygdala, an alternative model of MTLE which allows parallel experimental manipulation in the dentate gyrus, also trigger the induction of React-NSCs and provoke the disruption of the neurogenic niche resulting in impaired neurogenesis. These results highlight the sensitivity of NSCs to the surrounding neuronal circuit activity and demonstrate that the induction of React-NSCs and the disruption of the neurogenic niche are not due to the direct effect of KA in the hippocampus. These results also suggest that neurogenesis might be lost in the hippocampus of patients with MTLE. Indeed we provide results from human MTLE samples absence of cell proliferation, of neural stem cell-like cells and of neurogenesis.

11.
Cell Stem Cell ; 16(5): 488-503, 2015 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-25957904

RESUMEN

Adult hippocampal neurogenesis is believed to maintain a range of cognitive functions, many of which decline with age. We recently reported that radial neural stem cells (rNSCs) in the hippocampus undergo activation-dependent conversion into astrocytes, a mechanism that over time contributes to a reduction in the rNSC population. Here, we injected low and high levels of kainic acid (KA) in the dentate gyrus to assess whether neuronal hyperexcitation, a hallmark of epileptic disorders, could accelerate this conversion. At low levels of KA, generating epileptiform activity without seizures, we indeed found increased rNSC activation and conversion into astrocytes. At high levels, generating sustained epileptic seizures, however, we find that rNSCs divide symmetrically and that both mother and daughter cells convert into reactive astrocytes. Our results demonstrate that a threshold response for neuronal hyperexcitation provokes a dramatic shift in rNSC function, which impairs adult hippocampal neurogenesis in the long term.


Asunto(s)
Astrocitos/fisiología , Epilepsia/fisiopatología , Hipocampo/citología , Células-Madre Neurales/fisiología , Agitación Psicomotora/fisiopatología , Adulto , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Epilepsia/inducido químicamente , Humanos , Ácido Kaínico/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Neurogénesis
12.
Int J Dev Neurosci ; 31(7): 640-5, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23588197

RESUMEN

The generation of new neurons from neural stem cells (NSCs) throughout adult life in the mammalian brain is a biological process that fascinates scientists for its uniqueness and restorative potential. In the dentate gyrus (DG) of the hippocampus NSCs are able to self-renew and generate new granule cells and astrocytes through a complex and plastic mechanism that can be regulated by endogenous and exogenous cues at different levels. Unexpected recent findings suggest that the population of NSCs is heterogeneous in morphology and behavior. We herein explore the hypothesis that NSC heterogeneity and the neurogenic potential of the DG depends on their developmental origin. We provide an up-to-date picture of the process of neurogenesis in the adult hippocampus with an especial focus on NSCs and outline key unsolved aspects. Further, we discuss the origin of NSCs in the adult DG from a developmental perspective and explore the possibility of NSC heterogeneity being determined from early postnatal periods and being responsible for the neurogenic output of the DG in the long term.


Asunto(s)
Células Madre Adultas/fisiología , Hipocampo/citología , Hipocampo/fisiología , Neurogénesis/fisiología , Animales , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA