Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Nature ; 488(7412): 522-6, 2012 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-22854781

RESUMEN

Glioblastoma multiforme is the most common primary malignant brain tumour, with a median survival of about one year. This poor prognosis is due to therapeutic resistance and tumour recurrence after surgical removal. Precisely how recurrence occurs is unknown. Using a genetically engineered mouse model of glioma, here we identify a subset of endogenous tumour cells that are the source of new tumour cells after the drug temozolomide (TMZ) is administered to transiently arrest tumour growth. A nestin-ΔTK-IRES-GFP (Nes-ΔTK-GFP) transgene that labels quiescent subventricular zone adult neural stem cells also labels a subset of endogenous glioma tumour cells. On arrest of tumour cell proliferation with TMZ, pulse-chase experiments demonstrate a tumour re-growth cell hierarchy originating with the Nes-ΔTK-GFP transgene subpopulation. Ablation of the GFP+ cells with chronic ganciclovir administration significantly arrested tumour growth, and combined TMZ and ganciclovir treatment impeded tumour development. Thus, a relatively quiescent subset of endogenous glioma cells, with properties similar to those proposed for cancer stem cells, is responsible for sustaining long-term tumour growth through the production of transient populations of highly proliferative cells.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Dacarbazina/análogos & derivados , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Animales , Antineoplásicos Alquilantes/farmacología , Antineoplásicos Alquilantes/uso terapéutico , Proliferación Celular/efectos de los fármacos , Rastreo Celular , Dacarbazina/farmacología , Dacarbazina/uso terapéutico , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Ganciclovir/farmacología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Ratones , Ratones Transgénicos , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/patología , Temozolomida , Transgenes/genética
2.
Biol Psychiatry ; 95(1): 15-26, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-37423591

RESUMEN

BACKGROUND: Traumatic brain injury (TBI) is a debilitating neurological disorder caused by an impact to the head by an outside force. TBI results in persistent cognitive impairments, including fear generalization and the inability to distinguish between aversive and neutral stimuli. The mechanisms underlying fear generalization have not been fully elucidated, and there are no targeted therapeutics to alleviate this symptom of TBI. METHODS: To identify the neural ensembles mediating fear generalization, we utilized ArcCreERT2 × enhanced yellow fluorescent protein (EYFP) mice, which allow for activity-dependent labeling and quantification of memory traces. Mice were administered a sham surgery or the controlled cortical impact model of TBI. Mice were then administered a contextual fear discrimination paradigm and memory traces were quantified in numerous brain regions. In a separate group of mice, we tested if (R,S)-ketamine could decrease fear generalization and alter the corresponding memory traces in TBI mice. RESULTS: TBI mice exhibited increased fear generalization when compared with sham mice. This behavioral phenotype was paralleled by altered memory traces in the dentate gyrus, CA3, and amygdala, but not by alterations in inflammation or sleep. In TBI mice, (R,S)-ketamine facilitated fear discrimination, and this behavioral improvement was reflected in dentate gyrus memory trace activity. CONCLUSIONS: These data show that TBI induces fear generalization by altering fear memory traces and that this deficit can be improved with a single injection of (R,S)-ketamine. This work enhances our understanding of the neural basis of TBI-induced fear generalization and reveals potential therapeutic avenues for alleviating this symptom.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Ketamina , Ratones , Animales , Ketamina/farmacología , Hipocampo/metabolismo , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/metabolismo , Miedo , Encéfalo , Ratones Endogámicos C57BL
3.
bioRxiv ; 2023 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-36909465

RESUMEN

INTRODUCTION: Traumatic brain injury (TBI) is a debilitating neurological disorder caused by an impact to the head by an outside force. TBI results in persistent cognitive impairments, including fear generalization, the inability to distinguish between aversive and neutral stimuli. The mechanisms underlying fear generalization have not been fully elucidated, and there are no targeted therapeutics to alleviate this symptom of TBI. METHODS: To identify the neural ensembles mediating fear generalization, we utilized the ArcCreER T2 x enhanced yellow fluorescent protein (EYFP) mice, which allow for activity-dependent labeling and quantification of memory traces. Mice were administered a sham surgery or the controlled cortical impact (CCI) model of TBI. Mice were then administered a contextual fear discrimination (CFD) paradigm and memory traces were quantified in numerous brain regions. In a separate group of mice, we tested if ( R,S )-ketamine could decrease fear generalization and alter the corresponding memory traces in TBI mice. RESULTS: TBI mice exhibited increased fear generalization when compared with sham mice. This behavioral phenotype was paralleled by altered memory traces in the DG, CA3, and amygdala, but not by alterations in inflammation or sleep. In TBI mice, ( R,S )-ketamine facilitated fear discrimination and this behavioral improvement was reflected in DG memory trace activity. CONCLUSIONS: These data show that TBI induces fear generalization by altering fear memory traces, and that this deficit can be improved with a single injection of ( R,S )-ketamine. This work enhances our understanding of the neural basis of TBI-induced fear generalization and reveals potential therapeutic avenues for alleviating this symptom.

4.
J Neurosci ; 31(13): 4906-16, 2011 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-21451029

RESUMEN

Significant spontaneous recovery occurs after essentially all forms of serious brain injury, although the mechanisms underlying this recovery are unknown. Given that many forms of brain injury such as traumatic brain injury (TBI) induce hippocampal neurogenesis, we investigated whether these newly generated neurons might play a role in recovery. By modeling TBI in transgenic mice, we determined that injury-induced newly generated neurons persisted over time and elaborated extensive dendritic trees that stably incorporated themselves throughout all neuronal layers of the dentate gyrus. When we selectively ablated dividing stem/progenitors at the time of injury with ganciclovir in a nestin-HSV-TK transgenic model, we eliminated injury-induced neurogenesis and subsequently diminished the progenitor pool. Moreover, using hippocampal-specific behavioral tests, we demonstrated that only injured animals with neurogenesis ablated at the time of injury lost the ability to learn spatial memory tasks. These data demonstrate a functional role for adult neurogenesis after brain injury and offer compelling and testable therapeutic options that might enhance recovery.


Asunto(s)
Lesiones Encefálicas/genética , Trastornos del Conocimiento/genética , Cognición/fisiología , Neurogénesis/fisiología , Recuperación de la Función/fisiología , Animales , Lesiones Encefálicas/patología , Lesiones Encefálicas/fisiopatología , Trastornos del Conocimiento/patología , Trastornos del Conocimiento/fisiopatología , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Transgénicos , Factores de Tiempo
5.
Commun Biol ; 4(1): 1303, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34795427

RESUMEN

Polymorphisms in the apolipoprotein E (ApoE) gene confer a major genetic risk for the development of late-onset Alzheimer's disease (AD) and are predictive of outcome following traumatic brain injury (TBI). Alterations in adult hippocampal neurogenesis have long been associated with both the development of AD and recovery following TBI and ApoE is known to play a role in this process. In order to determine how ApoE might influence hippocampal injury-induced neurogenesis, we generated a conditional knockout system whereby functional ApoE from astrocytes was ablated prior to injury. While successfully ablating ApoE just prior to TBI in mice, we observed an attenuation in the development of the spines in the newborn neurons. Intriguingly, animals with a double-hit, i.e. injury and ApoE conditionally inactivated in astrocytes, demonstrated the most pronounced impairments in the hippocampal-dependent Morris water maze test, failing to exhibit spatial memory after both acquisition and reversal training trials. In comparison, conditional knockout mice without injury displayed impairments but only in the reversal phase of the test, suggesting accumulative effects of astrocytic ApoE deficiency and traumatic brain injury on AD-like phenotypes. Together, these findings demonstrate that astrocytic ApoE is required for functional injury-induced neurogenesis following traumatic brain injury.


Asunto(s)
Apolipoproteínas E/genética , Lesiones Traumáticas del Encéfalo/fisiopatología , Cognición , Neurogénesis , Neuronas/patología , Animales , Apolipoproteínas E/metabolismo , Astrocitos/metabolismo , Modelos Animales de Enfermedad , Femenino , Hipocampo/patología , Masculino , Ratones , Ratones Noqueados
6.
Dev Neurosci ; 32(5-6): 499-509, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21160162

RESUMEN

Brain remodeling occurs after all forms of brain injury, though the mechanisms underlying this phenomenon are mostly unknown. Neural stem and progenitor cells are one source of endogenous cells that may contribute to brain remodeling and subsequent recovery. In addition, certain populations of progenitors are particularly susceptible to injury, and their depletion may lead to the impairment of developmental processes that vary with age. We particularly focus on glial progenitors, which are more abundant postnatally and particularly susceptible to acquired brain injuries such as traumatic brain injury. We have recently characterized a novel transgenic mouse that expresses herpes thymidine kinase under the control of the neural-progenitor-specific nestin promoter and allows for temporally induced ablation of dividing progenitors. By genetically depleting dividing cortical progenitors at various times, we identify postnatal day 7 (P7) to P14 as a critical period for oligodendrogenesis. Targeted ablation of dividing progenitors during this window leads to cell-specific depletion of oligodendrocyte precursors expressing platelet-derived growth factor receptor-α and corresponding myelination and motor deficits. This modeling provides insight into how the age at which white matter injury occurs influences both injury severity and subsequent recovery.


Asunto(s)
Lesiones Encefálicas/patología , Oligodendroglía/patología , Células Madre/patología , Factores de Edad , Animales , Western Blotting , Modelos Animales de Enfermedad , Inmunohistoquímica , Ratones , Ratones Transgénicos , Microscopía Confocal
7.
PLoS One ; 15(3): e0229240, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32119690

RESUMEN

Various brain injuries lead to the activation of adult neural stem/progenitor cells in the mammalian hippocampus. Subsequent injury-induced neurogenesis appears to be essential for at least some aspects of the innate recovery in cognitive function observed following traumatic brain injury (TBI). It has previously been established that Apolipoprotein E (ApoE) plays a regulatory role in adult hippocampal neurogenesis, which is of particular interest as the presence of the human ApoE isoform ApoE4 leads to significant risk for the development of late-onset Alzheimer's disease, where impaired neurogenesis has been linked with disease progression. Moreover, genetically modified mice lacking ApoE or expressing the ApoE4 human isoform have been shown to impair adult hippocampal neurogenesis under normal conditions. Here, we investigate how controlled cortical impact (CCI) injury affects dentate gyrus development using hippocampal stereotactic injections of GFP-expressing retroviruses in wild-type (WT), ApoE-deficient and humanized (ApoE3 and ApoE4) mice. Infected adult-born hippocampal neurons were morphologically analyzed once fully mature, revealing significant attenuation of dendritic complexity and spine density in mice lacking ApoE or expressing the human ApoE4 allele, which may help inform how ApoE influences neurological diseases where neurogenesis is defective.


Asunto(s)
Apolipoproteína E3/metabolismo , Apolipoproteína E4/metabolismo , Apolipoproteínas E/deficiencia , Lesiones Traumáticas del Encéfalo/patología , Hipocampo/crecimiento & desarrollo , Retroviridae/genética , Animales , Apolipoproteína E3/genética , Apolipoproteína E4/genética , Lesiones Traumáticas del Encéfalo/genética , Lesiones Traumáticas del Encéfalo/metabolismo , Modelos Animales de Enfermedad , Femenino , Vectores Genéticos/administración & dosificación , Hipocampo/metabolismo , Humanos , Masculino , Ratones , Ratones Transgénicos , Neurogénesis , Neuronas/metabolismo
8.
Neuron ; 107(3): 552-565.e10, 2020 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-32502462

RESUMEN

The occurrence of dreaming during rapid eye movement (REM) sleep prompts interest in the role of REM sleep in hippocampal-dependent episodic memory. Within the mammalian hippocampus, the dentate gyrus (DG) has the unique characteristic of exhibiting neurogenesis persisting into adulthood. Despite their small numbers and sparse activity, adult-born neurons (ABNs) in the DG play critical roles in memory; however, their memory function during sleep is unknown. Here, we investigate whether young ABN activity contributes to memory consolidation during sleep using Ca2+ imaging in freely moving mice. We found that contextual fear learning recruits a population of young ABNs that are reactivated during subsequent REM sleep against a backdrop of overall reduced ABN activity. Optogenetic silencing of this sparse ABN activity during REM sleep alters the structural remodeling of spines on ABN dendrites and impairs memory consolidation. These findings provide a causal link between ABN activity during REM sleep and memory consolidation.


Asunto(s)
Condicionamiento Psicológico , Giro Dentado/fisiología , Consolidación de la Memoria/fisiología , Neuronas/fisiología , Sueño REM/fisiología , Animales , Calcio/metabolismo , Giro Dentado/citología , Electroencefalografía , Electromiografía , Miedo , Hipocampo , Aprendizaje , Ratones , Neurogénesis , Optogenética , Ritmo Teta
9.
J Neurosci ; 28(48): 12901-12, 2008 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-19036984

RESUMEN

It is becoming increasingly clear that brain injuries from a variety of causes stimulate neurogenesis within the hippocampus. It remains unclear, however, how robust this response may be and what primary cell types are involved. Here, using a controlled cortical impact model of traumatic brain injury on a previously characterized transgenic mouse line that expresses enhanced green fluorescent protein (eGFP) under the control of the nestin promoter, we demonstrate that it is the earliest type-1 quiescent progenitor cells that are induced to proliferate and migrate outside the subgranular layer of the dentate gyrus. This type-1 cell activation occurs at the same time that we observe adjacent but more differentiated doublecortin-expressing progenitors (type-2 cells) being eliminated. Also, although type-2 cells remain intact in the contralateral (uninjured) dentate gyrus, the type-1 cells there are also activated and result in increased numbers of the doublecortin-expressing type-2 cells. In addition, we have generated a novel mouse transgenic that expresses a modified version of the herpes simplex virus thymidine kinase along with eGFP that allows for the visualization and inducible ablation of early dividing progenitors by exposing them to ganciclovir. Using this transgenic in the context of traumatic brain injury, we demonstrate that these early progenitors are required for injury-induced remodeling to occur. This work suggests that injury-induced hippocampal remodeling following brain injury likely requires sustained activation of quiescent early progenitors.


Asunto(s)
Lesiones Encefálicas/metabolismo , Hipocampo/metabolismo , Proteínas de Filamentos Intermediarios/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/fisiología , Neuronas/metabolismo , Células Madre/metabolismo , Animales , Antivirales/farmacología , Biomarcadores/análisis , Biomarcadores/metabolismo , Lesiones Encefálicas/fisiopatología , Giro Dentado/citología , Giro Dentado/metabolismo , Modelos Animales de Enfermedad , Proteínas de Dominio Doblecortina , Ganciclovir/farmacología , Proteínas Fluorescentes Verdes/genética , Hipocampo/citología , Masculino , Ratones , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/metabolismo , Regeneración Nerviosa/genética , Nestina , Plasticidad Neuronal/genética , Neuropéptidos/metabolismo , Regiones Promotoras Genéticas/genética , Células Madre/citología , Timidina Quinasa/metabolismo
10.
eNeuro ; 5(4)2018.
Artículo en Inglés | MEDLINE | ID: mdl-30079373

RESUMEN

Adult hippocampal neurogenesis occurs throughout life and is believed to participate in cognitive functions such as learning and memory. A number of genes that regulate adult hippocampal neurogenesis have been identified, although most of these have been implicated in progenitor proliferation and survival, but not in the development into fully differentiated neurons. Among these genes, apolipoprotein E (ApoE) is particularly compelling because the human ApoE isoform E4 is a risk factor for the development of Alzheimer's disease, where hippocampal neurogenesis is reported to be dysfunctional. To investigate the effects of ApoE and its human isoforms on adult hippocampal neurogenesis and neuronal development, retroviruses carrying a GFP-expressing vector were injected into wild-type (WT), ApoE-deficient, and human targeted replacement (ApoE3 and ApoE4) mice to infect progenitors in the dentate gyrus and analyze the morphology of fully developed GFP-expressing neurons. Analysis of these adult-born neurons revealed significant decreases in the complexity of dendritic arborizations and spine density in ApoE-deficient mice compared with WT mice, as well as in ApoE4 mice compared with ApoE3. These findings demonstrate that ApoE deficiency and the ApoE4 human isoform both impair hippocampal neurogenesis and give insight into how ApoE may influence hippocampal-related neurological diseases.


Asunto(s)
Apolipoproteínas E/fisiología , Giro Dentado/fisiología , Neurogénesis/fisiología , Neuronas/patología , Neuronas/fisiología , Animales , Apolipoproteínas E/deficiencia , Giro Dentado/citología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neurogénesis/genética , Isoformas de Proteínas
11.
Brain Behav ; 7(9): e00796, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28948089

RESUMEN

INTRODUCTION: Significant enhancement of neurogenesis is known to occur in response to a variety of brain insults such as traumatic brain injury. Previous studies have demonstrated that injury-induced newborn neurons are required for hippocampus-dependent spatial learning and memory tasks like the Morris water maze, but not in contextual fear conditioning that requires both the hippocampus and amygdala. Recently, the dentate gyrus, where adult hippocampal neurogenesis occurs, has been implicated in processing information to form specific memory under specific environmental stimuli in a process known as pattern separation. METHODS: To test whether injury-induced newborn neurons facilitate pattern separation, hippocampus-dependent contextual fear discrimination was performed using delta-HSV-TK transgenic mice, which can temporally inhibit injury-induced neurogenesis under the control of ganciclovir. RESULTS: We observed that impaired neurogenesis enhanced the ability to distinguish aversive from naïve environments. In addition, this occurs most significantly following injury, but only in a context-dependent manner whereby the sequence of introducing the naïve environment from the aversive one affected the performance differentially. CONCLUSIONS: Temporal impairment of both baseline and injury-induced adult neurogenesis enhances performance in fear discrimination in a context-dependent manner.


Asunto(s)
Giro Dentado/fisiología , Discriminación en Psicología/fisiología , Miedo/fisiología , Neurogénesis/fisiología , Neuronas/fisiología , Animales , Giro Dentado/citología , Hipocampo/citología , Hipocampo/fisiología , Masculino , Memoria/fisiología , Ratones , Ratones Transgénicos , Neuronas/citología
12.
Mol Neurodegener ; 12(1): 64, 2017 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-28886753

RESUMEN

BACKGROUND: The molecular mechanism underlying progressive memory loss in Alzheimer's disease is poorly understood. Neurogenesis in the adult hippocampus is a dynamic process that continuously changes the dentate gyrus and is important for hippocampal plasticity, learning and memory. However, whether impairments in neurogenesis affect the hippocampal circuitry in a way that leads to memory deficits characteristic of Alzheimer's disease is unknown. Controversial results in that regard were reported in transgenic mouse models of amyloidosis. METHODS: Here, we conditionally ablated adult neurogenesis in APPswe/PS1ΔE9 mice by crossing these with mice expressing nestin-driven thymidine kinase (δ-HSV-TK). RESULTS: These animals show impairment in performance in contextual conditioning and pattern separation tasks following depletion of neurogenesis. Importantly, these deficits were not observed in age-matched APPswe/PS1ΔE9 or δ-HSV-TK mice alone. Furthermore, we show that cognitive deficits were accompanied by the upregulation of hyperphosphorylated tau in the hippocampus and in immature neurons specifically. Interestingly, we observed upregulation of the immediate early gene Zif268 (Egr-1) in the dentate gyrus, CA1 and CA3 regions of the hippocampus following learning in the neurogenesis-depleted δ-HSV-TK mice. This may suggest overactivation of hippocampal neurons in these areas following depletion of neurogenesis. CONCLUSIONS: These results imply that neurogenesis plays an important role in the regulation of inhibitory circuitry of the hippocampus. This study suggests that deficits in adult neurogenesis may contribute to cognitive impairments, tau hyperphosphorylation in new neurons and compromised hippocampal circuitry in Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Hipocampo/fisiopatología , Células-Madre Neurales/patología , Neurogénesis/fisiología , Animales , Ratones , Ratones Transgénicos
13.
Neuroscientist ; 22(1): 61-71, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25520428

RESUMEN

Partial recovery from brain injury due to trauma, hypoxia, or stroke, is ubiquitous and occurs largely through unknown mechanisms. It is now well accepted that injury enhances proliferation of quiescent stem and progenitor cells in specialized niches within the brain. However, whether this injury-induced neurogenesis contributes to recovery after brain injury remains controversial. Recent evidence suggests that hippocampal neural stem/precursor cell activation and subsequent neurogenesis are responsible for at least some aspects of spontaneous recovery following brain injury from a variety of causes. However, other aspects of injury-induced neurogenesis, including its contribution to adverse sequelae such as seizures, are still being investigated. The purpose of this review is to provide an overview of adult hippocampal neurogenesis and how it relates to injury and explain how current mouse technology is allowing for better understanding of whether manipulating this natural process might eventually help inform therapy following brain injury.


Asunto(s)
Lesiones Encefálicas/fisiopatología , Neurogénesis/fisiología , Animales , Encéfalo/fisiopatología , Humanos
14.
J Neurotrauma ; 33(4): 362-74, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25905575

RESUMEN

Partial recovery from even severe traumatic brain injury (TBI) is ubiquitous and occurs largely through unknown mechanisms. Recent evidence suggests that hippocampal neural stem/progenitor cell (NSPC) activation and subsequent neurogenesis are responsible for at least some aspects of spontaneous recovery following TBI. Apolipoprotein E (ApoE) regulates postnatal neurogenesis in the hippocampus and is therefore a putative mediator of injury-induced neurogenesis. Further, ApoE isoforms in humans are associated with different cognitive outcomes following TBI. To investigate the role of ApoE in injury-induced neurogenesis, we exposed wild-type, ApoE-deficient, and human ApoE isoform-specific (ApoE3 and ApoE4) transgenic mice crossed with nestin-green fluorescent protein (GFP) reporter mice to controlled cortical impact (CCI) and assessed progenitor activation at 2 d post-injury using unbiased stereology. GFP+ progenitor cells were increased by approximately 120% in the ipsilateral hippocampus in injured wild-type mice, compared with sham mice (p<0.01). Co-localization of GFP+ cells with bromodeoxyrudine (BrdU) to label dividing cells indicated increased proliferation of progenitors in the injured hippocampus (p<0.001). This proliferative injury response was absent in ApoE-deficient mice, as no increase in GFP+ cells was observed in the injured hippocampus, compared with sham mice, despite an overall increase in proliferation indicated by increased BrdU+ cells (86%; p<0.05). CCI-induced proliferation of GFP+ cells in both ApoE3 and ApoE4 mice but the overall response was attenuated in ApoE4 mice due to fewer GFP+ cells at baseline. We demonstrate that ApoE is required for injury-induced proliferation of NSPCs after experimental TBI, and that this response is influenced by human APOE genotype.


Asunto(s)
Apolipoproteínas E/fisiología , Lesiones Encefálicas/metabolismo , Hipocampo/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Animales , Lesiones Encefálicas/patología , Femenino , Hipocampo/patología , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Células-Madre Neurales/patología , Células Madre/metabolismo , Células Madre/patología
15.
Mech Dev ; 116(1-2): 187-92, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12128222

RESUMEN

In a subtraction designed to identify transcripts accompanying mesodermal lineage specification in mouse ES differentiation cultures, we identified a gene encoding a two LIM-domain protein which we named heart LIM protein (Hlp). Hlp is most closely related to thymus LIM protein, and these two genes comprise a new gene family related to the cysteine-rich protein (CRP) gene family. In the embryo, Hlp expression is primarily restricted to the developing heart. In situ hybridization showed expression at E7.75 in the paired heart-forming primordia prior to linear heart-tube formation. At E8.5, strong expression is detected in the heart, with equal expression in both heart chambers. Hlp expression is detected in both myocardium and endocardium, and in vascular endothelium. Later in fetal development low levels of Hlp expression are detected outside the heart, including dorsal root ganglia and the spinal cord. In the adult, Hlp is expressed at highest levels in the heart, and at lower levels in the brain, skeletal muscle and aorta. Hlp expression is unchanged in hypertrophic hearts induced by aortic constriction. These data suggest a role for the two LIM-domain gene Hlp in the very earliest stages of heart differentiation and development.


Asunto(s)
Corazón Fetal/embriología , Corazón Fetal/metabolismo , Miocardio/metabolismo , Proteínas Nucleares/genética , Proteínas , Secuencia de Aminoácidos , Animales , Endotelio Vascular/embriología , Endotelio Vascular/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ , Proteínas con Dominio LIM , Ratones , Datos de Secuencia Molecular , Filogenia , Homología de Secuencia de Aminoácido , Distribución Tisular
16.
PLoS One ; 10(2): e0118793, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25714524

RESUMEN

Traumatic brain injury (TBI) is ubiquitous and effective treatments for it remain supportive largely due to uncertainty over how endogenous repair occurs. Recently, we demonstrated that hippocampal injury-induced neurogenesis is one mechanism underlying endogenous repair following TBI. Donepezil is associated with increased hippocampal neurogenesis and has long been known to improve certain aspects of cognition following many types of brain injury through unknown mechanisms. By coupling donepezil therapy with temporally regulated ablation of injury-induced neurogenesis using nestin-HSV transgenic mice, we investigated whether the pro-cognitive effects of donepezil following injury might occur through increasing neurogenesis. We demonstrate that donepezil itself enhances neurogenesis and improves cognitive function following TBI, even when injury-induced neurogenesis was inhibited. This suggests that the therapeutic effects of donepezil in TBI occur separately from its effects on neurogenesis.


Asunto(s)
Lesiones Encefálicas/fisiopatología , Indanos/farmacología , Trastornos de la Memoria/tratamiento farmacológico , Neurogénesis/efectos de los fármacos , Piperidinas/farmacología , Aprendizaje Espacial/efectos de los fármacos , Animales , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/patología , Giro Dentado/efectos de los fármacos , Modelos Animales de Enfermedad , Donepezilo , Femenino , Ganciclovir/análogos & derivados , Ganciclovir/farmacología , Indanos/administración & dosificación , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Transgénicos , Piperidinas/administración & dosificación , Valganciclovir
17.
J Neurotrauma ; 32(11): 753-64, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25290253

RESUMEN

Although a myriad of pathological responses contribute to traumatic brain injury (TBI), cerebral dysfunction has been closely linked to cell death mechanisms. A number of therapeutic strategies have been studied in an attempt to minimize or ameliorate tissue damage; however, few studies have evaluated the inherent protective capacity of the brain. Endogenous neural stem/progenitor cells (NSPCs) reside in distinct brain regions and have been shown to respond to tissue damage by migrating to regions of injury. Until now, it remained unknown whether these cells have the capacity to promote endogenous repair. We ablated NSPCs in the subventricular zone to examine their contribution to the injury microenvironment after controlled cortical impact (CCI) injury. Studies were performed in transgenic mice expressing the herpes simplex virus thymidine kinase gene under the control of the nestin(δ) promoter exposed to CCI injury. Two weeks after CCI injury, mice deficient in NSPCs had reduced neuronal survival in the perilesional cortex and fewer Iba-1-positive and glial fibrillary acidic protein-positive glial cells but increased glial hypertrophy at the injury site. These findings suggest that the presence of NSPCs play a supportive role in the cortex to promote neuronal survival and glial cell expansion after TBI injury, which corresponds with improvements in motor function. We conclude that enhancing this endogenous response may have acute protective roles after TBI.


Asunto(s)
Lesiones Encefálicas/metabolismo , Microambiente Celular/fisiología , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Células-Madre Neurales/metabolismo , Animales , Lesiones Encefálicas/patología , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Ventrículos Laterales/citología , Ventrículos Laterales/metabolismo , Ratones , Ratones Transgénicos , Células-Madre Neurales/patología , Neurogénesis/fisiología
18.
Nat Commun ; 3: 1253, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23212382

RESUMEN

Different places may share common features, but are coded by distinct populations of CA3 neurons in the hippocampus. Here we show that chemical or genetic suppression of adult neurogenesis in the hippocampus impairs this population-based coding of similar (but not dissimilar) contexts. These data provide a neural basis for impaired spatial discrimination following ablation of adult neurogenesis, and support the proposal that adult neurogenesis regulates the efficiency of a pattern separation process in the hippocampus.


Asunto(s)
Región CA3 Hipocampal/fisiología , Neurogénesis/fisiología , Alquilantes/farmacología , Animales , Región CA3 Hipocampal/efectos de los fármacos , Condicionamiento Psicológico/efectos de los fármacos , Condicionamiento Psicológico/fisiología , Dacarbazina/análogos & derivados , Dacarbazina/farmacología , Discriminación en Psicología/efectos de los fármacos , Discriminación en Psicología/fisiología , Ganciclovir/farmacología , Hipocampo/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neurogénesis/efectos de los fármacos , Temozolomida
19.
Genesis ; 41(4): 147-53, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15789426

RESUMEN

The use of mouse gene targeting to study molecules important in neural development is oftentimes impaired by early embryonic lethality. In order to address later roles for such molecules, specifically in neural stem cells, we generated transgenic mice that express both the tetracycline-inducible molecule rtTA-M2 and GFP under the control of the neural precursor specific form of nestin. Developmental analysis of these mice demonstrates that GFP expression is exclusive to the neural tube. Adult expression of GFP is seen only in known areas of adult neurogenesis, namely, the subventricular zone and the dentate gyrus. When crossed with a second transgenic mouse (TetOp-Cre) that expresses the Cre recombinase under the control of the tetracycline responsive promotor, we demonstrate temporal induction of Cre in bigenic animals exposed to doxycycline. We further demonstrate the feasibility of this approach by using the ROSA-26 reporter mouse to mediate recombination in neural precursor cells.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Integrasas/biosíntesis , Neuronas/fisiología , Células Madre/fisiología , Animales , Antibacterianos/farmacología , Sistema Nervioso Central/embriología , Genes Reporteros , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Tetraciclina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA