Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 986
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
Glia ; 72(9): 1693-1706, 2024 09.
Artículo en Inglés | MEDLINE | ID: mdl-38852127

RESUMEN

Astrocytes that reside in superficial (SL) and deep cortical layers have distinct molecular profiles and morphologies, which may underlie specific functions. Here, we demonstrate that the production of SL and deep layer (DL) astrocyte populations from neural progenitor cells in the mouse is temporally regulated. Lineage tracking following in utero and postnatal electroporation with PiggyBac (PB) EGFP and birth dating with EdU and FlashTag, showed that apical progenitors produce astrocytes during late embryogenesis (E16.5) that are biased to the SL, while postnatally labeled (P0) astrocytes are biased to the DL. In contrast, astrocytes born during the predominantly neurogenic window (E14.5) showed a random distribution in the SL and DL. Of interest, E13.5 astrocytes birth dated at E13.5 with EdU showed a lower layer bias, while FT labeling of apical progenitors showed no bias. Finally, examination of the morphologies of "biased" E16.5- and P0-labeled astrocytes demonstrated that E16.5-labeled astrocytes exhibit different morphologies in different layers, while P0-labeled astrocytes do not. Differences based on time of birth are also observed in the molecular profiles of E16.5 versus P0-labeled astrocytes. Altogether, these results suggest that the morphological, molecular, and positional diversity of cortical astrocytes is related to their time of birth from ventricular/subventricular zone progenitors.


Asunto(s)
Astrocitos , Corteza Cerebral , Células-Madre Neurales , Animales , Astrocitos/metabolismo , Astrocitos/citología , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología , Ratones , Ratones Transgénicos , Femenino , Animales Recién Nacidos , Regulación del Desarrollo de la Expresión Génica , Transcriptoma , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Ventrículos Cerebrales/citología , Ratones Endogámicos C57BL
2.
Dev Growth Differ ; 66(5): 329-337, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38894655

RESUMEN

Microglia colonize the brain starting on embryonic day (E) 9.5 in mice, and their population increases with development. We have previously demonstrated that some microglia are derived from intraventricular macrophages, which frequently infiltrate the pallium at E12.5. To address how the infiltration of intraventricular macrophages is spatiotemporally regulated, histological analyses detecting how these cells associate with the surrounding cells at the site of infiltration into the pallial surface are essential. Using two-photon microscopy-based in vivo imaging, we demonstrated that most intraventricular macrophages adhere to the ventricular surface. This is a useful tool for imaging intraventricular macrophages maintaining their original position, but this method cannot be used for observing deeper brain regions. Meanwhile, we found that conventional cryosection-based and naked pallial slice-based observation resulted in unexpected detachment from the ventricular surface of intraventricular macrophages and their mislocation, suggesting that previous histological analyses might have failed to determine their physiological number and location in the ventricular space. To address this, we sought to establish a methodological preparation that enables us to delineate the structure and cellular interactions when intraventricular macrophages infiltrate the pallium. Here, we report that brain slices pretreated with agarose-embedding maintained adequate density and proper positioning of intraventricular macrophages on the ventricular surface. This method also enabled us to perform the immunostaining. We believe that this is helpful for conducting histological analyses to elucidate the mechanisms underlying intraventricular macrophage infiltration into the pallium and their cellular properties, leading to further understanding of the process of microglial colonization into the developing brain.


Asunto(s)
Encéfalo , Macrófagos , Animales , Macrófagos/citología , Ratones , Encéfalo/embriología , Encéfalo/citología , Microglía/citología , Microglía/metabolismo , Ventrículos Cerebrales/embriología , Ventrículos Cerebrales/citología
3.
J Neurosci ; 41(15): 3301-3306, 2021 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-33597270

RESUMEN

Heterogeneity is defined as the quality or state of being diverse in character or content. This article summarizes the natural progression from my studies, reported in the first issue of the Journal of Neuroscience, that identified molecular heterogeneity in precursor cells of the developing primate cerebral cortex to the current state in which differences defined at the molecular, cellular, circuit, and systems levels are building data encyclopedias. The emphasis on heterogeneity has impacted many contributors in the field of developmental neuroscience, who have led a quest to determine the extent to which there is diversity, when it appears developmentally, and what heritable and nonheritable factors mediate nervous system assembly and function. Since the appearance of the article on progenitor cell heterogeneity in the inaugural issue of the Journal of Neuroscience, there have been continuous advances in technologies and data analytics that are contributing to a much better understanding of the origins of neurobiological and behavioral heterogeneity.


Asunto(s)
Ventrículos Cerebrales/citología , Células-Madre Neurales/fisiología , Neurogénesis , Neuroglía/fisiología , Animales , Ventrículos Cerebrales/crecimiento & desarrollo , Ventrículos Cerebrales/fisiología , Humanos , Células-Madre Neurales/citología , Neuroglía/citología
4.
Cereb Cortex ; 31(4): 2139-2155, 2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33279961

RESUMEN

Microglial cells make extensive contacts with neural precursor cells (NPCs) and affiliate with vasculature in the developing cerebral cortex. But how vasculature contributes to cortical histogenesis is not yet fully understood. To better understand functional roles of developing vasculature in the embryonic rat cerebral cortex, we investigated the temporal and spatial relationships between vessels, microglia, and NPCs in the ventricular zone. Our results show that endothelial cells in developing cortical vessels extend numerous fine processes that directly contact mitotic NPCs and microglia; that these processes protrude from vessel walls and are distinct from tip cell processes; and that microglia, NPCs, and vessels are highly interconnected near the ventricle. These findings demonstrate the complex environment in which NPCs are embedded in cortical proliferative zones and suggest that developing vasculature represents a source of signaling with the potential to broadly influence cortical development. In summary, cortical histogenesis arises from the interplay among NPCs, microglia, and developing vasculature. Thus, factors that impinge on any single component have the potential to change the trajectory of cortical development and increase susceptibility for altered neurodevelopmental outcomes.


Asunto(s)
Ventrículos Cerebrales/irrigación sanguínea , Ventrículos Cerebrales/embriología , Neocórtex/irrigación sanguínea , Neocórtex/embriología , Neurogénesis/fisiología , Neuroinmunomodulación/fisiología , Animales , Ventrículos Cerebrales/citología , Desarrollo Embrionario/fisiología , Femenino , Microglía/fisiología , Neocórtex/citología , Células-Madre Neurales/fisiología , Embarazo , Ratas
5.
Bioessays ; 42(3): e1900186, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32078177

RESUMEN

The brain ventricular system is a series of connected cavities, filled with cerebrospinal fluid (CSF), that forms within the vertebrate central nervous system (CNS). The hollow neural tube is a hallmark of the chordate CNS, and a closed neural tube is essential for normal development. Development and function of the ventricular system is examined, emphasizing three interdigitating components that form a functional system: ventricle walls, CSF fluid properties, and activity of CSF constituent factors. The cellular lining of the ventricle both can produce and is responsive to CSF. Fluid properties and conserved CSF components contribute to normal CNS development. Anomalies of the CSF/ventricular system serve as diagnostics and may cause CNS disorders, further highlighting their importance. This review focuses on the evolution and development of the brain ventricular system, associated function, and connected pathologies. It is geared as an introduction for scholars with little background in the field.


Asunto(s)
Ventrículos Cerebrales/crecimiento & desarrollo , Ventrículos Cerebrales/metabolismo , Líquido Cefalorraquídeo/metabolismo , Animales , Evolución Biológica , Encefalopatías/metabolismo , Ventrículos Cerebrales/citología , Presión del Líquido Cefalorraquídeo/fisiología , Proteínas del Líquido Cefalorraquídeo/metabolismo , Cilios/metabolismo , Epitelio/crecimiento & desarrollo , Epitelio/metabolismo , Humanos , Cinética , Tubo Neural/citología , Tubo Neural/crecimiento & desarrollo , Tubo Neural/metabolismo , Transducción de Señal
6.
Cereb Cortex ; 30(7): 4092-4109, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32108222

RESUMEN

Even after birth, neuronal production continues in the ventricular-subventricular zone (V-SVZ) and hippocampus in many mammals. The immature new neurons ("neuroblasts") migrate and then mature at their final destination. In humans, neuroblast production and migration toward the neocortex and the olfactory bulb (OB) occur actively only for a few months after birth and then sharply decline with age. However, the precise spatiotemporal profiles and fates of postnatally born neurons remain unclear due to methodological limitations. We previously found that common marmosets, small nonhuman primates, share many features of V-SVZ organization with humans. Here, using marmosets injected with thymidine analogue(s) during various postnatal periods, we demonstrated spatiotemporal changes in neurogenesis during development. V-SVZ progenitor proliferation and neuroblast migration toward the OB and neocortex sharply decreased by 4 months, most strikingly in a V-SVZ subregion from which neuroblasts migrated toward the neocortex. Postnatally born neurons matured within a few months in the OB and hippocampus but remained immature until 6 months in the neocortex. While neurogenic activity was sustained for a month after birth, the distribution and/or differentiation diversity was more restricted in 1-month-born cells than in the neonatal-born population. These findings shed light on distinctive features of postnatal neurogenesis in primates.


Asunto(s)
Proliferación Celular , Hipocampo/crecimiento & desarrollo , Ventrículos Laterales/crecimiento & desarrollo , Neocórtex/crecimiento & desarrollo , Células-Madre Neurales/citología , Neurogénesis , Bulbo Olfatorio/crecimiento & desarrollo , Animales , Encéfalo/citología , Encéfalo/crecimiento & desarrollo , Callithrix , Movimiento Celular , Ventrículos Cerebrales/citología , Ventrículos Cerebrales/crecimiento & desarrollo , Hipocampo/citología , Ventrículos Laterales/citología , Neocórtex/citología , Bulbo Olfatorio/citología , Análisis Espacio-Temporal
7.
Glia ; 68(2): 435-450, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31626379

RESUMEN

Postnatal subventricular zone (SVZ) neural stem cells generate forebrain glia, namely astrocytes and oligodendrocytes. The cues necessary for this process are unclear, despite this phase of brain development being pivotal in forebrain gliogenesis. Galectin-3 (Gal-3) is increased in multiple brain pathologies and thereby regulates astrocyte proliferation and inflammation in injury. To study the function of Gal-3 in inflammation and gliogenesis, we carried out functional studies in mouse. We overexpressed Gal-3 with electroporation and using immunohistochemistry surprisingly found no inflammation in the healthy postnatal SVZ. This allowed investigation of inflammation-independent effects of Gal-3 on gliogenesis. Loss of Gal-3 function via knockdown or conditional knockout reduced gliogenesis, whereas Gal-3 overexpression increased it. Gal-3 overexpression also increased the percentage of striatal astrocytes generated by the SVZ but decreased the percentage of oligodendrocytes. These novel findings were further elaborated with multiple analyses demonstrating that Gal-3 binds to the bone morphogenetic protein receptor one alpha (BMPR1α) and increases bone morphogenetic protein (BMP) signaling. Conditional knockout of BMPR1α abolished the effect of Gal-3 overexpression on gliogenesis. Gain-of-function of Gal-3 is relevant in pathological conditions involving the human forebrain, which is particularly vulnerable to hypoxia/ischemia during perinatal gliogenesis. Hypoxic/ischemic injury induces astrogliosis, inflammation and cell death. We show that Gal-3 immunoreactivity was increased in the perinatal human SVZ and striatum after hypoxia/ischemia. Our findings thus show a novel inflammation-independent function for Gal-3; it is necessary for gliogenesis and when increased in expression can induce astrogenesis via BMP signaling.


Asunto(s)
Astrocitos/metabolismo , Galectina 3/metabolismo , Ventrículos Laterales/citología , Neuroglía/metabolismo , Animales , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Ventrículos Cerebrales/citología , Regulación de la Expresión Génica , Isquemia/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Oligodendroglía/metabolismo
8.
J Anat ; 236(2): 334-350, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31670387

RESUMEN

The ventricular layer of the spinal cord is remodelled during embryonic development and ultimately forms the ependymal cell lining of the adult central canal, which retains neural stem cell potential. This anatomical transformation involves the process of dorsal collapse; however, accompanying changes in tissue organisation and cell behaviour as well as the precise origin of cells contributing to the central canal are not well understood. Here, we describe sequential localised cell rearrangements which accompany the gradual attrition of the spinal cord ventricular layer during development. This includes local breakdown of the pseudostratified organisation of the dorsal ventricular layer prefiguring dorsal collapse and evidence for a new phenomenon, ventral dissociation, during which the ventral-most floor plate cells separate from a subset that are retained around the central canal. Using cell proliferation markers and cell-cycle reporter mice, we further show that following dorsal collapse, ventricular layer attrition involves an overall reduction in cell proliferation, characterised by an intriguing increase in the percentage of cells in G1/S. In contrast, programmed cell death does not contribute to ventricular layer remodelling. By analysing transcript and protein expression patterns associated with key signalling pathways, we provide evidence for a gradual decline in ventral sonic hedgehog activity and an accompanying ventral expansion of initial dorsal bone morphogenetic protein signalling, which comes to dominate the forming the central canal lining. This study identifies multiple steps that may contribute to spinal cord ventricular layer attrition and adds to increasing evidence for the heterogeneous origin of the spinal cord ependymal cell population, which includes cells from the floor plate and the roof plate as well as ventral progenitor domains.


Asunto(s)
Proliferación Celular/fisiología , Ventrículos Cerebrales/citología , Médula Espinal/citología , Animales , Apoptosis/fisiología , Proteínas Morfogenéticas Óseas/metabolismo , Ciclo Celular/fisiología , Ventrículos Cerebrales/metabolismo , Epéndimo/citología , Epéndimo/metabolismo , Proteínas Hedgehog/metabolismo , Ratones , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal/fisiología , Médula Espinal/metabolismo
9.
Nature ; 497(7449): 369-73, 2013 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-23615612

RESUMEN

Postnatal/adult neural stem cells (NSCs) within the rodent subventricular zone (SVZ; also called subependymal zone) generate doublecortin (Dcx)(+) neuroblasts that migrate and integrate into olfactory bulb circuitry. Continuous production of neuroblasts is controlled by the SVZ microenvironmental niche. It is generally thought that enhancing the neurogenic activities of endogenous NSCs may provide needed therapeutic options for disease states and after brain injury. However, SVZ NSCs can also differentiate into astrocytes. It remains unclear whether there are conditions that favour astrogenesis over neurogenesis in the SVZ niche, and whether astrocytes produced there have different properties compared with astrocytes produced elsewhere in the brain. Here we show in mice that SVZ-generated astrocytes express high levels of thrombospondin 4 (Thbs4), a secreted homopentameric glycoprotein, in contrast to cortical astrocytes, which express low levels of Thbs4. We found that localized photothrombotic/ischaemic cortical injury initiates a marked increase in Thbs4(hi) astrocyte production from the postnatal SVZ niche. Tamoxifen-inducible nestin-creER(tm)4 lineage tracing demonstrated that it is these SVZ-generated Thbs4(hi) astrocytes, and not Dcx(+) neuroblasts, that home-in on the injured cortex. This robust post-injury astrogenic response required SVZ Notch activation modulated by Thbs4 via direct Notch1 receptor binding and endocytosis to activate downstream signals, including increased Nfia transcription factor expression important for glia production. Consequently, Thbs4 homozygous knockout mice (Thbs4(KO/KO)) showed severe defects in cortical-injury-induced SVZ astrogenesis, instead producing cells expressing Dcx migrating from SVZ to the injury sites. These alterations in cellular responses resulted in abnormal glial scar formation after injury, and significantly increased microvascular haemorrhage into the brain parenchyma of Thbs4(KO/KO) mice. Taken together, these findings have important implications for post-injury applications of endogenous and transplanted NSCs in the therapeutic setting, as well as disease states where Thbs family members have important roles.


Asunto(s)
Astrocitos/citología , Astrocitos/metabolismo , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Ventrículos Cerebrales/citología , Receptor Notch1/metabolismo , Trombospondinas/metabolismo , Animales , Linaje de la Célula , Movimiento Celular , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Cicatriz/metabolismo , Cicatriz/patología , Proteína Doblecortina , Endocitosis , Ratones , Ratones Noqueados , Factores de Transcripción NFI/metabolismo , Células-Madre Neurales/citología , Neuroglía/citología , Neuroglía/metabolismo , Neuroglía/patología , Transducción de Señal , Trombospondinas/deficiencia , Trombospondinas/genética
10.
Cell Mol Life Sci ; 75(6): 1027-1041, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29018869

RESUMEN

Originating from ectodermal epithelium, radial glial cells (RGCs) retain apico-basolateral polarity and comprise a pseudostratified epithelial layer in the developing cerebral cortex. The apical endfeet of the RGCs faces the fluid-filled ventricles, while the basal processes extend across the entire cortical span towards the pial surface. RGC functions are largely dependent on this polarized structure and the molecular components that define it. In this review, we will dissect existing molecular evidence on RGC polarity establishment and during cerebral cortex development and provide our perspective on the remaining key questions.


Asunto(s)
Polaridad Celular , Corteza Cerebral/metabolismo , Ectodermo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas del Tejido Nervioso/genética , Neuroglía/metabolismo , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/ultraestructura , Uniones Adherentes/metabolismo , Uniones Adherentes/ultraestructura , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Corteza Cerebral/citología , Corteza Cerebral/crecimiento & desarrollo , Ventrículos Cerebrales/citología , Ventrículos Cerebrales/crecimiento & desarrollo , Ventrículos Cerebrales/metabolismo , Ectodermo/citología , Ectodermo/crecimiento & desarrollo , Embrión de Mamíferos , Epitelio/crecimiento & desarrollo , Epitelio/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/ultraestructura , Humanos , Proteínas del Tejido Nervioso/metabolismo , Neuroglía/citología , Piamadre/citología , Piamadre/crecimiento & desarrollo , Piamadre/metabolismo
11.
Glia ; 66(12): 2659-2672, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30338559

RESUMEN

Nuclear factor-kappaB (NF-κB) is activated in neural progenitor cells in the developing murine cerebral cortex during the neurogenic phase, when it acts to prevent premature neuronal differentiation. Here we show that NF-κB activation continues in mouse neocortical neural progenitor cells during the neurogenic-to-gliogenic switch. Blockade of endogenous NF-κB activity during neocortical gliogenesis leads to the formation of supernumerary committed gliogenic progenitors and premature glial cell differentiation. Conversely, forced NF-κB activation during the neocortical neurogenic-to-gliogenic transition causes delayed gliogenic commitment and decreased astroglial gene expression. NF-κB activation continues in neocortical gliogenic progenitors following commitment and is important to inhibit the differentiation of oligodendrocyte precursor cells and to maintain persistent expression of glial fibrillary acidic protein in maturing astrocytes. These results reveal a number of previously uncharacterized roles for NF-κB during different phases of neocortical gliogenesis and identify NF-κB as an inhibitor of early oligodendrocyte development in the cerebral cortex.


Asunto(s)
Corteza Cerebral , Regulación del Desarrollo de la Expresión Génica/genética , FN-kappa B/metabolismo , Neurogénesis/genética , Neuroglía/fisiología , Animales , Animales Recién Nacidos , Diferenciación Celular/fisiología , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/embriología , Corteza Cerebral/crecimiento & desarrollo , Ventrículos Cerebrales/citología , Ventrículos Cerebrales/embriología , Ventrículos Cerebrales/crecimiento & desarrollo , Factor Neurotrófico Ciliar/farmacología , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteína Ácida Fibrilar de la Glía/metabolismo , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Antígeno Ki-67/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , FN-kappa B/genética , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/fisiología
12.
J Neurosci ; 36(29): 7786-801, 2016 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-27445154

RESUMEN

UNLABELLED: As neural structures grow in size and increase metabolic demand, the CNS vasculature undergoes extensive growth, remodeling, and maturation. Signals from neural tissue act on endothelial cells to stimulate blood vessel ingression, vessel patterning, and acquisition of mature brain vascular traits, most notably the blood-brain barrier. Using mouse genetic and in vitro approaches, we identified retinoic acid (RA) as an important regulator of brain vascular development via non-cell-autonomous and cell-autonomous regulation of endothelial WNT signaling. Our analysis of globally RA-deficient embryos (Rdh10 mutants) points to an important, non-cell-autonomous function for RA in the development of the vasculature in the neocortex. We demonstrate that Rdh10 mutants have severe defects in cerebrovascular development and that this phenotype correlates with near absence of endothelial WNT signaling, specifically in the cerebrovasculature, and substantially elevated expression of WNT inhibitors in the neocortex. We show that RA can suppress the expression of WNT inhibitors in neocortical progenitors. Analysis of vasculature in non-neocortical brain regions suggested that RA may have a separate, cell-autonomous function in brain endothelial cells to inhibit WNT signaling. Using both gain and loss of RA signaling approaches, we show that RA signaling in brain endothelial cells can inhibit WNT-ß-catenin transcriptional activity and that this is required to moderate the expression of WNT target Sox17. From this, a model emerges in which RA acts upstream of the WNT pathway via non-cell-autonomous and cell-autonomous mechanisms to ensure the formation of an adequate and stable brain vascular plexus. SIGNIFICANCE STATEMENT: Work presented here provides novel insight into important yet little understood aspects of brain vascular development, implicating for the first time a factor upstream of endothelial WNT signaling. We show that RA is permissive for cerebrovascular growth via suppression of WNT inhibitor expression in the neocortex. RA also functions cell-autonomously in brain endothelial cells to modulate WNT signaling and its downstream target, Sox17. The significance of this is although endothelial WNT signaling is required for neurovascular development, too much endothelial WNT signaling, as well as overexpression of its target Sox17, are detrimental. Therefore, RA may act as a "brake" on endothelial WNT signaling and Sox17 to ensure normal brain vascular development.


Asunto(s)
Encéfalo/citología , Ventrículos Cerebrales/citología , Regulación del Desarrollo de la Expresión Génica/genética , Receptor alfa de Ácido Retinoico/metabolismo , Tretinoina/metabolismo , Vía de Señalización Wnt/fisiología , Factores de Edad , Oxidorreductasas de Alcohol/deficiencia , Oxidorreductasas de Alcohol/genética , Animales , Encéfalo/embriología , Diferenciación Celular , Células Cultivadas , Ventrículos Cerebrales/embriología , Embrión de Mamíferos , Células Endoteliales/metabolismo , Efrinas/genética , Efrinas/metabolismo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Receptor alfa de Ácido Retinoico/genética , Tretinoina/farmacología , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
13.
J Neurosci ; 36(4): 1203-10, 2016 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-26818508

RESUMEN

We demonstrated previously that Pannexin 1 (Panx1), an ion and metabolite channel, promotes the growth and proliferation of ventricular zone (VZ) neural precursor cells (NPCs) in vitro. To investigate its role in vivo, we used floxed Panx1 mice in combination with viruses to delete Panx1 in VZ NPCs and to track numbers of Panx1-null and Panx1-expressing VZ NPCs over time. Two days after virus injection, Panx1-null cells were less abundant than Panx1-expressing cells, suggesting that Panx1 is required for the maintenance of VZ NPCs. We also investigated the effect of Panx1 deletion in VZ NPCs after focal cortical stroke via photothrombosis. Panx1 is essential for maintaining elevated VZ NPC numbers after stroke. In contrast, Panx1-null NPCs were more abundant than Panx1-expressing NPCs in the peri-infarct cortex. Together, these findings suggest that Panx1 plays an important role in NPC maintenance in the VZ niche in the naive and stroke brain and could be a key target for improving NPC survival in the peri-infarct cortex. SIGNIFICANCE STATEMENT: Here, we demonstrate that Pannexin 1 (Panx1) maintains a consistent population size of neural precursor cells in the ventricular zone, both in the healthy brain and in the context of stroke. In contrast, Panx1 appears to be detrimental to the survival of neural precursor cells that surround damaged cortical tissue in the stroke brain. This suggests that targeting Panx1 in the peri-infarct cortex, in combination with other therapies, could improve cell survival around the injury site.


Asunto(s)
Infarto Cerebral/patología , Ventrículos Cerebrales/citología , Conexinas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/fisiología , Neurogénesis/fisiología , Análisis de Varianza , Animales , Caspasa 3/metabolismo , Recuento de Células , Supervivencia Celular/fisiología , Conexinas/genética , Modelos Animales de Enfermedad , Proteínas de Dominio Doblecortina , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Antígeno Ki-67/metabolismo , Ratones , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas del Tejido Nervioso/genética , Neurogénesis/genética , Neuropéptidos/metabolismo , Accidente Cerebrovascular/complicaciones
14.
Biochem Biophys Res Commun ; 493(1): 751-757, 2017 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-28859977

RESUMEN

Due to their widely known therapeutic benefits, mesenchymal stem cells have been proposed as a novel treatment option for a wide range of diseases including Alzheimer's disease. To maximize these benefits, critical factors such as delivery route, cell viability, and cell migration must be accounted for. Out of the various delivery routes to the brain, the intracerebroventricular (ICV) route stands out due to the widespread distribution that can occur via cerebrospinal fluid flow. The major objective of this present study was to observe how altering cell concentration influences the migration and viability of human umbilical cord blood derived-mesenchymal stem cells (hUCB-MSCs), delivered via ICV injection, in the brains of wild-type (WT) mice. C3H/C57 WT mice were divided into three groups and were injected with 1 × 105 hUCB-MSCs suspended in varying volumes: high (3 µl), middle (5 µl), and low (7 µl) concentrations, respectively. Lowering the concentration increased the migratory capabilities and elevated the viability of hUCB-MSCs. These results suggest that cell concentration can affect the physiological state of hUCB-MSCs, and thus the extent of therapeutic efficacy that can be achieved.


Asunto(s)
Movimiento Celular/fisiología , Supervivencia Celular/fisiología , Ventrículos Cerebrales/citología , Ventrículos Cerebrales/fisiología , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Animales , Recuento de Células , Células Cultivadas , Humanos , Inyecciones Intraventriculares/métodos , Ratones , Ratones Endogámicos C3H
15.
Proc Natl Acad Sci U S A ; 111(29): 10726-31, 2014 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-25002477

RESUMEN

Postnatal neurogenesis in mammals is confined to restricted brain regions, including the subventricular zone (SVZ). In rodents, the SVZ is a lifelong source of new neurons fated to migrate to the olfactory bulb (OB), where the majority become GABAergic interneurons. The plastic capacity of neonatal and adult SVZ stem/progenitor cells is still largely unknown. By overexpressing the transcription factor Fezf2, a powerful master gene specifying the phenotype of glutamatergic subcerebral projecting neurons, we investigated whether the fate of postnatally generated SVZ neurons can be altered. Following lentiviral delivery of Fezf2 in the neonatal and adult SVZ niche, we showed that ectopic Fezf2 expression is sufficient to redirect the fate of SVZ stem cells. Thus, based on in vivo and in vitro experiments, we provide evidence that numerous Fezf2-positive OB neurons expressed glutamatergic pyramidal cell molecular markers instead of developing a GABAergic identity. Overexpression of Fezf2 had no effect on transit-amplifying progenitors or neuroblasts but was restricted to neural stem cells. Fezf2-respecified neurons bore features of pyramidal cells, exhibiting a larger cell body and a more elaborate dendritic tree, compared with OB granule cells. Patch-clamp recordings further indicated that Fezf2-respecified neurons had synaptic properties and a firing pattern reminiscent of a pyramidal cell-like phenotype. Together, the results demonstrate that neonatal and adult SVZ stem cells retain neuronal fate plasticity.


Asunto(s)
Diferenciación Celular , Corteza Cerebral/citología , Ventrículos Cerebrales/citología , Proteínas de Unión al ADN/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Potenciales de Acción , Envejecimiento/metabolismo , Animales , Linaje de la Célula , Dendritas/metabolismo , Neuronas GABAérgicas/citología , Neuronas GABAérgicas/metabolismo , Glutamatos/metabolismo , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Fenotipo , Células Piramidales/citología , Células Piramidales/metabolismo , Sinapsis/metabolismo
16.
J Neurosci ; 35(11): 4528-39, 2015 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-25788671

RESUMEN

Adult neural stem cells reside in specialized niches. In the ventricular-subventricular zone (V-SVZ), quiescent neural stem cells (qNSCs) become activated (aNSCs), and generate transit amplifying cells (TACs), which give rise to neuroblasts that migrate to the olfactory bulb. The vasculature is an important component of the adult neural stem cell niche, but whether vascular cells in neurogenic areas are intrinsically different from those elsewhere in the brain is unknown. Moreover, the contribution of pericytes to the neural stem cell niche has not been defined. Here, we describe a rapid FACS purification strategy to simultaneously isolate primary endothelial cells and pericytes from brain microregions of nontransgenic mice using CD31 and CD13 as surface markers. We compared the effect of purified vascular cells from a neurogenic (V-SVZ) and non-neurogenic brain region (cortex) on the V-SVZ stem cell lineage in vitro. Endothelial and pericyte diffusible signals from both regions differentially promote the proliferation and neuronal differentiation of qNSCs, aNSCs, and TACs. Unexpectedly, diffusible cortical signals had the most potent effects on V-SVZ proliferation and neurogenesis, highlighting the intrinsic capacity of non-neurogenic vasculature to support stem cell behavior. Finally, we identify PlGF-2 as an endothelial-derived mitogen that promotes V-SVZ cell proliferation. This purification strategy provides a platform to define the functional and molecular contribution of vascular cells to stem cell niches and other brain regions under different physiological and pathological states.


Asunto(s)
Células Madre Adultas/fisiología , Linaje de la Célula/fisiología , Ventrículos Cerebrales/fisiología , Endotelio Vascular/fisiología , Células-Madre Neurales/fisiología , Neurogénesis/fisiología , Animales , Proliferación Celular/fisiología , Células Cultivadas , Ventrículos Cerebrales/citología , Endotelio Vascular/citología , Masculino , Ratones
17.
J Neurochem ; 139(2): 245-255, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27607605

RESUMEN

Class III phosphoinositide 3-kinase (PIK3C3 or mammalian vacuolar protein sorting 34 homolog, Vps34) regulates vesicular trafficking, autophagy, and nutrient sensing. Recently, we reported that PIK3C3 is expressed in mouse cerebral cortex throughout the developmental process, especially at early embryonic stage. We thus examined the role of PIK3C3 in the development of the mouse cerebral cortex. Acute silencing of PIK3C3 with in utero electroporation method caused positional defects of excitatory neurons during corticogenesis. Time-lapse imaging revealed that the abnormal positioning was at least partially because of the reduced migration velocity. When PIK3C3 was silenced in cortical neurons in one hemisphere, axon extension to the contralateral hemisphere was also delayed. These aberrant phenotypes were rescued by RNAi-resistant PIK3C3. Notably, knockdown of PIK3C3 did not affect the cell cycle of neuronal progenitors and stem cells at the ventricular zone. Taken together, PIK3C3 was thought to play a crucial role in corticogenesis through the regulation of excitatory neuron migration and axon extension. Meanwhile, when we performed comparative genomic hybridization on a patient with specific learning disorders, a 107 Kb-deletion was identified on 18q12.3 (nt. 39554147-39661206) that encompasses exons 5-23 of PIK3C3. Notably, the above aberrant migration and axon growth phenotypes were not rescued by the disease-related truncation mutant (172 amino acids) lacking the C-terminal kinase domain. Thus, functional defects of PIK3C3 might impair corticogenesis and relate to the pathophysiology of specific learning disorders and other neurodevelopmental disorders. Acute knockdown of Class III phosphoinositide 3-kinase (PIK3C3) evokes migration defects of excitatory neurons during corticogenesis. PIK3C3-knockdown also disrupts axon outgrowth, but not progenitor proliferation in vivo. Involvement of PIK3C3 in neurodevelopmental disorders might be an interesting future subject since a deletion mutation in PIK3C3 was detected in a patient with specific learning disorders (SLD).


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido/genética , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Encéfalo/enzimología , Encéfalo/crecimiento & desarrollo , Discapacidades para el Aprendizaje/genética , Animales , Axones , Encéfalo/embriología , Movimiento Celular/genética , Corteza Cerebral/embriología , Corteza Cerebral/enzimología , Corteza Cerebral/crecimiento & desarrollo , Ventrículos Cerebrales/citología , Ventrículos Cerebrales/enzimología , Ventrículos Cerebrales/crecimiento & desarrollo , Niño , Exones/genética , Femenino , Eliminación de Gen , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Humanos , Pruebas de Inteligencia , Discapacidades para el Aprendizaje/psicología , Ratones , Células-Madre Neurales , Hibridación de Ácido Nucleico , Embarazo , Interferencia de ARN
18.
Eur J Neurosci ; 43(11): 1474-85, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26946195

RESUMEN

Primitive neural stem cells (pNSCs) are the earliest NSCs to appear in the developing forebrain. They persist into the adult forebrain where they can generate all cells in the neural lineage and therefore hold great potential for brain regeneration. Thus, pNSCs are an ideal population to target to promote endogenous NSC activation. pNSCs can be isolated from the periventricular region as leukaemia inhibitory factor-responsive cells, and comprise a rare population in the adult mouse brain. We hypothesized that the pup periventricular region gives rise to more clonal pNSC-derived neurospheres but that pup-derived pNSCs are otherwise comparable to adult-derived pNSCs, and can be used to identify selective markers and activators of endogenous pNSCs. We tested the self-renewal ability, differentiation capacity and gene expression profile of pup-derived pNSCs and found them each to be comparable to adult-derived pNSCs, including being GFAP(-) , nestin(mid) , Oct4(+) . Next, we used pup pNSCs to test pharmacological compounds to activate pNSCs to promote endogenous brain repair. We hypothesized that pNSCs could be activated by targeting the cell surface proteins C-Kit and ErbB2, which were enriched in pNSCs relative to definitive NSCs (dNSCs) in an in vitro screen. C-Kit and ErbB2 signalling inhibition had distinct effects on pNSCs and dNSCs in vitro, and when infused directly into the adult brain in vivo. Targeted activation of pNSCs with C-Kit and ErbB2 modulation is a valuable strategy to activate the earliest cell in the neural lineage to contribute to endogenous brain regeneration.


Asunto(s)
Encéfalo/fisiología , Células-Madre Neurales/fisiología , Animales , Astrocitos/metabolismo , Astrocitos/fisiología , Encéfalo/citología , Encéfalo/metabolismo , Diferenciación Celular , Células Cultivadas , Ventrículos Cerebrales/citología , Expresión Génica , Proteína Ácida Fibrilar de la Glía/metabolismo , Inmunohistoquímica , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/metabolismo , Ratones , Nestina/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Neuronas/fisiología , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Oligodendroglía/metabolismo , Oligodendroglía/fisiología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptor ErbB-2/metabolismo , Factores de Transcripción SOXB1/metabolismo , beta Catenina/metabolismo
19.
J Neuroinflammation ; 13: 7, 2016 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-26747276

RESUMEN

BACKGROUND: Demyelination and axonal degeneration, hallmarks of multiple sclerosis (MS), are associated with the central nervous system (CNS) inflammation facilitated by C-X-C motif chemokine 12 (CXCL12) chemokine. Both in MS and in experimental autoimmune encephalomyelitis (EAE), the deleterious CNS inflammation has been associated with upregulation of CXCL12 expression in the CNS. We investigated the expression dynamics of CXCL12 in the CNS with progression of clinical EAE and following spontaneous recovery, with a focus on CXCL12 expression in the hippocampal neurogenic dentate gyrus (DG) and in the corpus callosum (CC) of spontaneously recovered mice, and its potential role in promoting the endogenous myelin/neuronal repair capacity. METHODS: CNS tissue sections from mice with different clinical EAE phases or following spontaneous recovery and in vitro differentiated adult neural stem cell cultures were analyzed by immunofluorescent staining and confocal imaging for detecting and enumerating neuronal progenitor cells (NPCs) and oligodendrocyte precursor cells (OPCs) and for expression of CXCL12. RESULTS: Our expression dynamics analysis of CXCL12 in the CNS with EAE progression revealed elevated CXCL12 expression in the DG and CC, which persistently increases following spontaneous recovery even though CNS inflammation has subsided. Correspondingly, the numbers of NPCs and OPCs in the DG and CC, respectively, of EAE-recovered mice increased compared to that of naïve mice (NPCs, p < 0.0001; OPCs, p < 0.00001) or mice with active disease (OPCs, p < 0.0005). Notably, about 30 % of the NPCs and unexpectedly also OPCs (~50 %) express CXCL12, and their numbers in DG and CC, respectively, are higher in EAE-recovered mice compared with naïve mice and also compared with mice with ongoing clinical EAE (CXCL12(+) NPCs, p < 0.005; CXCL12(+) OPCs, p < 0.0005). Moreover, a significant proportion (>20 %) of the CXCL12(+) NPCs and OPCs co-express the CXCL12 receptor, CXCR4, and their numbers significantly increase with recovery from EAE not only relative to naïve mice (p < 0.0002) but also to mice with ongoing EAE (p < 0.004). CONCLUSIONS: These data link CXCL12 expression in the DG and CC of EAE-recovering mice to the promotion of neuro/oligodendrogenesis generating CXCR4(+) CXCL12(+) neuronal and oligodendrocyte progenitor cells endowed with intrinsic neuro/oligondendroglial differentiation potential. These findings highlight the post-CNS-inflammation role of CXCL12 in augmenting the endogenous myelin/neuronal repair capacity in MS-like disease, likely via CXCL12/CXCR4 autocrine signaling.


Asunto(s)
Sistema Nervioso Central/metabolismo , Quimiocina CXCL12/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Vaina de Mielina/metabolismo , Recuperación de la Función/fisiología , Animales , Recuento de Células , Diferenciación Celular , Células Cultivadas , Ventrículos Cerebrales/citología , Quimiocina CXCL12/farmacología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Proteínas de Dominio Doblecortina , Encefalomielitis Autoinmune Experimental/inducido químicamente , Regulación de la Expresión Génica/inmunología , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/metabolismo , Proteína Proteolipídica de la Mielina/inmunología , Proteína Proteolipídica de la Mielina/toxicidad , Células-Madre Neurales/metabolismo , Neuronas/patología , Neuropéptidos/metabolismo , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/toxicidad , Cicatrización de Heridas/fisiología
20.
Acta Anaesthesiol Scand ; 60(5): 579-87, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26822861

RESUMEN

BACKGROUND: Developmental neurotoxicity of ketamine, an N-methyl-D-aspartate receptor antagonist, must be considered due to its widespread uses for sedation/analgesia/anesthesia in pediatric and obstetric settings. Dose-dependent effects of ketamine on cellular proliferation in the neurogenic regions of rat fetal cortex [ventricular zone (VZ) and subventricular zone (SVZ)] were investigated in this in vivo study. METHODS: Timed-pregnant Sprague-Dawley rats at embryonic day 17 (E17) were given with different doses of ketamine intraperitoneally (0, 1, 2, 10, 20, 40, and 100 mg/kg). Proliferating cells in the rat fetal brains were labeled by injecting 100 mg/kg of 5-bromo-2'-deoxyuridine (BrdU) intraperitoneally. BrdU-labeled cells were detected by immunostaining methods. The numbers of BrdU-positive cells in VZ and SVZ of rat fetal cortex were employed to quantify proliferation in the developing rat cortex. RESULTS: Ketamine dose-dependently reduced the number of BrdU-positive cells in VZ (P < 0.001) and SVZ (P < 0.001) of the rat fetal cortex. SVZ showed greater susceptibility to ketamine-induced reduction of proliferation in rat fetal cortex, occurring even at clinically relevant doses (2 mg/kg). CONCLUSION: These data suggest that exposure to ketamine during embryogenesis can dose-dependently inhibit the cellular proliferation in neurogenic regions of the rat fetal cortex.


Asunto(s)
Anestésicos Disociativos/toxicidad , Proliferación Celular/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/embriología , Desarrollo Embrionario/efectos de los fármacos , Ketamina/toxicidad , Neurogénesis/efectos de los fármacos , Animales , Encéfalo/efectos de los fármacos , Encéfalo/embriología , Recuento de Células , Ventrículos Cerebrales/citología , Ventrículos Cerebrales/efectos de los fármacos , Ventrículos Cerebrales/embriología , Relación Dosis-Respuesta a Droga , Femenino , Ventrículos Laterales/citología , Ventrículos Laterales/efectos de los fármacos , Ventrículos Laterales/embriología , Embarazo , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA