Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
J Neurosci ; 39(50): 9967-9988, 2019 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-31685650

RESUMEN

New neurons, referred to as neuroblasts, are continuously generated in the ventricular-subventricular zone of the brain throughout an animal's life. These neuroblasts are characterized by their unique potential for proliferation, formation of chain-like cell aggregates, and long-distance and high-speed migration through the rostral migratory stream (RMS) toward the olfactory bulb (OB), where they decelerate and differentiate into mature interneurons. The dynamic changes of ultrastructural features in postnatal-born neuroblasts during migration are not yet fully understood. Here we report the presence of a primary cilium, and its ultrastructural morphology and spatiotemporal dynamics, in migrating neuroblasts in the postnatal RMS and OB. The primary cilium was observed in migrating neuroblasts in the postnatal RMS and OB in male and female mice and zebrafish, and a male rhesus monkey. Inhibition of intraflagellar transport molecules in migrating neuroblasts impaired their ciliogenesis and rostral migration toward the OB. Serial section transmission electron microscopy revealed that each migrating neuroblast possesses either a pair of centrioles or a basal body with an immature or mature primary cilium. Using immunohistochemistry, live imaging, and serial block-face scanning electron microscopy, we demonstrate that the localization and orientation of the primary cilium are altered depending on the mitotic state, saltatory migration, and deceleration of neuroblasts. Together, our results highlight a close mutual relationship between spatiotemporal regulation of the primary cilium and efficient chain migration of neuroblasts in the postnatal brain.SIGNIFICANCE STATEMENT Immature neurons (neuroblasts) generated in the postnatal brain have a mitotic potential and migrate in chain-like cell aggregates toward the olfactory bulb. Here we report that migrating neuroblasts possess a tiny cellular protrusion called a primary cilium. Immunohistochemical studies with zebrafish, mouse, and monkey brains suggest that the presence of the primary cilium in migrating neuroblasts is evolutionarily conserved. Ciliogenesis in migrating neuroblasts in the rostral migratory stream is suppressed during mitosis and promoted after cell cycle exit. Moreover, live imaging and 3D electron microscopy revealed that ciliary localization and orientation change during saltatory movement of neuroblasts. Our results reveal highly organized dynamics in maturation and positioning of the primary cilium during neuroblast migration that underlie saltatory movement of postnatal-born neuroblasts.


Asunto(s)
Movimiento Celular/fisiología , Cilios/ultraestructura , Ventrículos Laterales/ultraestructura , Células-Madre Neurales/ultraestructura , Neuronas/ultraestructura , Bulbo Olfatorio/ultraestructura , Animales , Femenino , Macaca mulatta , Masculino , Ratones , Pez Cebra
2.
Stem Cells ; 34(10): 2574-2586, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27299504

RESUMEN

In the mouse brain, neuroblasts generated in the subventricular zone (SVZ) migrate to the olfactory bulb (OB) through the rostral migratory stream (RMS). Although the RMS is not present in the human brain, a migratory pathway that is organized around a ventricular cavity that reaches the OB has been reported. A similar cavity, the lateral ventricle extension (LVE), is found in the adult guinea pig brain. Therefore, we analyzed cytoarchitecture, proliferative activity and precursor cell migration in the SVZ and LVE of 1-, 6- and 12-month-old guinea pigs. In young animals, we used confocal spectral and transmission electron microscopy to identify neuroblasts, astrocytes, and progenitor cells in the SVZ and LVE. Analysis of peroxidase diffusion demonstrated that the LVE was a continuous cavity lined by ependymal cells and surrounded by neuroblasts. Precursor cells were mostly located in the SVZ and migrated from the SVZ to the OB through the LVE. Finally, analysis of 6- and 12-month-old guinea pigs revealed that the LVE was preserved in older animals; however, the number of neurogenic cells was significantly reduced. Consequently, we propose that the guinea pig brain may be used as a new neurogenic model with increased similarity to humans, given that the LVE connects the LV with the OB, as has been described in humans, and that the LVE works a migratory pathway. Stem Cells 2016;34:2574-2586.


Asunto(s)
Envejecimiento/fisiología , Movimiento Celular , Ventrículos Laterales/citología , Neuronas/citología , Animales , Proliferación Celular , Forma de la Célula , Cobayas , Ventrículos Laterales/ultraestructura , Masculino
3.
Adv Exp Med Biol ; 913: 397-402, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27796901

RESUMEN

It is well proved already that neurogenesis does take place in mammals' brain, including human brain. However, neurogenesis by itself is not able to compensate for brain tissue loss in serious neurological diseases, such as stroke, brain trauma or neurodegenerative disorders. Recent evidences show that neural stem cell niches are present not only in classical locations, such as subventricularor subgranular zones, but in other areas as well, including tissues contiguous to the brain (meninges and choroid plexus).In this chapter we revise the relationship of neural stem cells with interstitial cells (mainly telocytes), which we think is significant, and we describe what is known about the juxtacerebral tissue neurogenesis potential.


Asunto(s)
Plexo Coroideo/fisiología , Meninges/fisiología , Regeneración Nerviosa/fisiología , Células-Madre Neurales/fisiología , Nicho de Células Madre/fisiología , Telocitos/fisiología , Animales , Lesiones Traumáticas del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/rehabilitación , Plexo Coroideo/ultraestructura , Hipocampo/fisiología , Hipocampo/ultraestructura , Humanos , Ventrículos Laterales/fisiología , Ventrículos Laterales/ultraestructura , Meninges/ultraestructura , Microscopía Electrónica de Transmisión , Células-Madre Neurales/ultraestructura , Neurogénesis/fisiología , Ratas , Accidente Cerebrovascular/patología , Rehabilitación de Accidente Cerebrovascular , Telocitos/ultraestructura
4.
J Appl Toxicol ; 35(7): 737-51, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25256750

RESUMEN

Ochratoxin A (OTA), a mycotoxin that was discovered as a secondary metabolite of the fungal species Aspergillus and Penicillium, is a common contaminant in food and animal feed. This mycotoxin has been described as teratogenic, carcinogenic, genotoxic, immunotoxic and has been proven a potent neurotoxin. Other authors have previously reported the effects of OTA in different structures of the central nervous system as well as in some neurogenic regions. However, the impact of OTA exposure in the subventricular zone (SVZ) has not been assessed yet. To elucidate whether OTA affects neural precursors of the mouse SVZ we investigated, in vitro and in vivo, the effects of OTA exposure on the SVZ and on the neural precursors obtained from this neurogenic niche. In this work, we prove the cumulative effect of OTA exposure on proliferation, differentiation and depletion of neural stem cells cultured from the SVZ. In addition, we corroborated these results in vivo by immunohistochemistry and electron microscopy. As a result, we found a significant alteration in the proliferation process, which was evidenced by a decrease in the number of 5-bromo-2-deoxyuridine-positive cells and glial cells, as well as, a significant decrease in the number of neuroblasts in the SVZ. To summarize, in this study we demonstrate how OTA could be a threat to the developing and the adult SVZ through its impact in cell viability, proliferation and differentiation in a dose-dependent manner.


Asunto(s)
Ventrículos Laterales/efectos de los fármacos , Micotoxinas/toxicidad , Ocratoxinas/toxicidad , Animales , Astrocitos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Ventrículos Laterales/patología , Ventrículos Laterales/ultraestructura , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica , Células-Madre Neurales/efectos de los fármacos , Neuroglía/efectos de los fármacos
5.
Glia ; 62(5): 790-803, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24677590

RESUMEN

Neurogenesis persists in the adult subventricular zone (SVZ) of the mammalian brain. During aging, the SVZ neurogenic capacity undergoes a progressive decline, which is attributed to a decrease in the population of neural stem cells (NSCs). However, the behavior of the NSCs that remain in the aged brain is not fully understood. Here we performed a comparative ultrastructural study of the SVZ niche of 2-month-old and 24-month-old male C57BL/6 mice, focusing on the NSC population. Using thymidine-labeling, we showed that residual NSCs in the aged SVZ divide less frequently than those in young mice. We also provided evidence that ependymal cells are not newly generated during senescence, as others studies suggest. Remarkably, both astrocytes and ependymal cells accumulated a high number of intermediate filaments and dense bodies during aging, resembling reactive cells. A better understanding of the changes occurring in the neurogenic niche during aging will allow us to develop new strategies for fighting neurological disorders linked to senescence.


Asunto(s)
Envejecimiento/fisiología , Astrocitos/fisiología , Epéndimo/citología , Epéndimo/fisiología , Ventrículos Laterales/citología , Ventrículos Laterales/fisiología , Animales , Astrocitos/ultraestructura , Diferenciación Celular/fisiología , Proliferación Celular , Epéndimo/ultraestructura , Ventrículos Laterales/ultraestructura , Masculino , Ratones , Ratones Endogámicos C57BL , Células-Madre Neurales/fisiología , Células-Madre Neurales/ultraestructura , Neurogénesis/fisiología
6.
J Neurosci ; 32(34): 11511-23, 2012 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-22915098

RESUMEN

Hydrocephalus formation is a frequent complication of neuropathological insults associated with neuroinflammation. However, the mechanistic role of neuroinflammation in hydrocephalus development is unclear. We have investigated the function of the proinflammatory acting inhibitor of κB kinase (IKK)/nuclear factor κB (NF-κB) signaling system in neuroinflammatory processes and generated a novel mouse model that allows conditional activation of the IKK/NF-κB system in astrocytes. Remarkably, NF-κB activation in astrocytes during early postnatal life results in hydrocephalus formation and additional defects in brain development. NF-κB activation causes global neuroinflammation characterized by a strong, astrocyte-specific expression of proinflammatory NF-κB target genes as well as a massive infiltration and activation of macrophages. In this animal model, hydrocephalus formation is specifically induced during a critical time period of early postnatal development, in which IKK/NF-κB-induced neuroinflammation interferes with ependymal ciliogenesis. Our findings demonstrate for the first time that IKK/NF-κB activation is sufficient to induce hydrocephalus formation and provides a potential mechanistic explanation for the frequent association of neuroinflammation and hydrocephalus formation during brain development, namely impairment of ependymal cilia formation. Therefore, our study might open up new perspectives for the treatment of certain types of neonatal and childhood hydrocephalus associated with hemorrhages and infections.


Asunto(s)
Encefalitis/etiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Glioma Subependimario/etiología , Hidrocefalia , Quinasa I-kappa B/metabolismo , Factores de Edad , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Astrocitos/enzimología , Encéfalo/enzimología , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas , Corteza Cerebral/citología , Quimiocinas/genética , Quimiocinas/metabolismo , Proteínas del Sistema Complemento/genética , Proteínas del Sistema Complemento/metabolismo , Modelos Animales de Enfermedad , Doxiciclina/administración & dosificación , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/genética , Glioma Subependimario/patología , Humanos , Hidrocefalia/complicaciones , Hidrocefalia/enzimología , Hidrocefalia/patología , Quinasa I-kappa B/genética , Proteínas I-kappa B/metabolismo , Ventrículos Laterales/crecimiento & desarrollo , Ventrículos Laterales/patología , Ventrículos Laterales/ultraestructura , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Análisis por Micromatrices , Microscopía Electrónica de Rastreo , Inhibidor NF-kappaB alfa , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Estadísticas no Paramétricas , Factor de Transcripción ReIA/metabolismo , Quinasa de Factor Nuclear kappa B
7.
Dev Neurosci ; 34(4): 299-309, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22831917

RESUMEN

Much work has focused on the possible contribution of adult hippocampal neurogenesis to neuropsychiatric diseases. The hippocampal subgranular zone and the other stem cell-containing neurogenic niche, the subventricular zone (SVZ), share several cytological features and are regulated by some of the same molecular mechanisms. However, very little is known about the SVZ in neuropsychiatric disorders. This is important since it surrounds the lateral ventricles and in schizophrenia ventricular enlargement frequently follows forebrain nuclei shrinkage. Also, adult neurogenesis has been implicated in pharmacotherapy for affective disorders and many of the molecules associated with neuropsychiatric disorders affect SVZ biology. To assess the neurogenic niche, we examined material from 60 humans (Stanley Collection) and characterized the cytoarchitecture of the SVZ and ependymal layer in age-, sex- and post mortem interval-matched controls, and patients diagnosed with schizophrenia, bipolar illness, and depression (n = 15 each). There is a paucity of post mortem brains available for study in these diseases, so to maximize the number of possible parameters examined here, we quantified individual sections rather than a large series. Previous work showed that multiple sclerosis is associated with increased width of the hypocellular gap, a cell-sparse region that typifies the human SVZ. Statistically there were no differences between disease groups and controls in the width of the hypocellular gap or in the density of cells in the hypocellular gap. Because ventricular enlargement in schizophrenia may disrupt ependymal cells, we quantified them, but observed no difference between diagnostic groups and controls. There are significant differences in the prevalence of neuropsychiatric illness between the sexes. Therefore, we looked for male versus female differences, but did not observe any in the parameters quantified. We next turned to a finer spatial resolution and asked if there were differences amongst the disease groups in dorsal ventral subdivisions of the SVZ. Similar to when we treated the SVZ as a whole, we did not find such differences. However, compared to the dorsal SVZ, the ventral SVZ had a wider hypocellular gap and more ependymal cells in all four groups. In contrast, cell density was similar in dorsal ventral subregions of the SVZ hypocellular gap. These results show that though there are regional differences in the SVZ in humans, neuropsychiatric disorders do not seem to alter several fundamental histological features of this adult neurogenic zone.


Asunto(s)
Trastorno Bipolar/patología , Núcleo Caudado/ultraestructura , Trastorno Depresivo/patología , Epéndimo/ultraestructura , Hipocampo/ultraestructura , Ventrículos Laterales/ultraestructura , Putamen/ultraestructura , Esquizofrenia/patología , Adulto , Astrocitos/ultraestructura , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neurogénesis , Neuronas/ultraestructura , Especificidad de Órganos , Células Madre/ultraestructura , Adulto Joven
8.
J Neurosci ; 28(14): 3804-13, 2008 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-18385338

RESUMEN

The subventricular zone (SVZ) of the adult mouse brain is a narrow stem cell niche that lies along the length of the lateral wall of the lateral ventricles. The SVZ supports neurogenesis throughout adulthood; however, with increasing age, the ventral SVZ deteriorates and only the dorsolateral SVZ remains neurogenic. Associated with the elderly dorsolateral SVZ, we reported previously an increased number of astrocytes interposed within the adjacent ependymal lining. Here, we show that astrocytes integrated within the ependyma are dividing, BrdU-labeled astrocytes that share cellular adherens with neighboring ependymal cells. By tracking BrdU-labeled astrocytes over time, we observed that, as they incorporated within the ependyma, they took on antigenic and morphologic characteristics of ependymal cells, suggesting a novel form of SVZ-supported "regenerative" repair in the aging brain. A similar form of SVZ-mediated ependyma repair was also observed in young mice after mild ependymal cell denudation with low dosages of neuraminidase. Together, this work identifies a novel non-neuronal mechanism of regenerative repair by the adult SVZ.


Asunto(s)
Células Madre Adultas/fisiología , Envejecimiento/patología , Epéndimo/lesiones , Epéndimo/fisiopatología , Ventrículos Laterales/citología , Células Madre Adultas/ultraestructura , Factores de Edad , Animales , Astrocitos/fisiología , Astrocitos/ultraestructura , Encéfalo/anatomía & histología , Bromodesoxiuridina/metabolismo , Recuento de Células/métodos , Relación Dosis-Respuesta a Droga , Epéndimo/efectos de los fármacos , Epéndimo/ultraestructura , Ventrículos Laterales/ultraestructura , Masculino , Ratones , Microscopía Confocal/métodos , Microscopía Electrónica/métodos , Proteínas del Tejido Nervioso/metabolismo , Neuraminidasa/efectos adversos
9.
J Neurosci ; 28(48): 12887-900, 2008 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-19036983

RESUMEN

Primary cilia are important sites of signal transduction involved in a wide range of developmental and postnatal functions. Proteolytic processing of the transcription factor Gli3, for example, occurs in primary cilia, and defects in intraflagellar transport (IFT), which is crucial for the maintenance of primary cilia, can lead to severe developmental defects and diseases. Here we report an essential role of primary cilia in forebrain development. Uncovered by N-ethyl-N-nitrosourea-mutagenesis, cobblestone is a hypomorphic allele of the IFT gene Ift88, in which Ift88 mRNA and protein levels are reduced by 70-80%. cobblestone mutants are distinguished by subpial heterotopias in the forebrain. Mutants show both severe defects in the formation of dorsomedial telencephalic structures, such as the choroid plexus, cortical hem and hippocampus, and also a relaxation of both dorsal-ventral and rostral-caudal compartmental boundaries. These defects phenocopy many of the abnormalities seen in the Gli3 mutant forebrain, and we show that Gli3 proteolytic processing is reduced, leading to an accumulation of the full-length activator isoform. In addition, we observe an upregulation of canonical Wnt signaling in the neocortex and in the caudal forebrain. Interestingly, the ultrastructure and morphology of ventricular cilia in the cobblestone mutants remains intact. Together, these results indicate a critical role for ciliary function in the developing forebrain.


Asunto(s)
Corteza Cerebral/anomalías , Corteza Cerebral/metabolismo , Cilios/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Supresoras de Tumor/genética , Animales , Corteza Cerebral/ultraestructura , Cilios/ultraestructura , Epéndimo/metabolismo , Epéndimo/ultraestructura , Femenino , Factores de Transcripción de Tipo Kruppel/genética , Ventrículos Laterales/anomalías , Ventrículos Laterales/metabolismo , Ventrículos Laterales/ultraestructura , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Neuronas/ultraestructura , Péptido Hidrolasas/metabolismo , Prosencéfalo/anomalías , Prosencéfalo/metabolismo , Prosencéfalo/ultraestructura , Proteínas Supresoras de Tumor/metabolismo , Proteínas Wnt/metabolismo , Proteína Gli3 con Dedos de Zinc
10.
J Anat ; 213(3): 259-65, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18624828

RESUMEN

The microvascular architecture of developing lateral ventricle choroid plexus was investigated by corrosion casting and scanning electron microscopy in human fetuses aged 20 gestational weeks. The areas with different microvascular patterns corresponded to the particular parts of the mature plexus: anterior part, glomus, posterior part, the villous fringe and the free margin. In the posterior part, densely packed parallel arterioles and venules were surrounded by sheath-like capillary networks. Other areas contained compact capillary plexuses of the primary villi: the most prominent, protruding basket- and leaf-shaped plexuses were observed in the villous fringe, whilst less numerous and smaller plexuses occurred in the anterior part and glomus. The capillaries of the plexuses had a large diameter and sinusoidal dilations, and showed the presence of occasional short, blind sprouts indicative of angiogenesis. Short anastomoses between arterioles supplying the plexuses and venules draining them were only rarely observed. In the upper area of the choroid plexus, the superior choroidal vein was surrounded by a capillary network forming small, glomerular or rosette-shapes plexuses. The free margin of the choroid plexus was characterized by flat, multiple, arcade-like capillary loops. The general vascular architecture of the human choroid plexus at 20 gestational weeks seems to be similar to that of postnatal/mature plexus, still lacking, however, the complex vascular plexuses of the secondary villi.


Asunto(s)
Plexo Coroideo/irrigación sanguínea , Plexo Coroideo/embriología , Ventrículos Laterales/embriología , Arteriolas/embriología , Arteriolas/ultraestructura , Capilares/embriología , Capilares/ultraestructura , Plexo Coroideo/ultraestructura , Molde por Corrosión , Femenino , Edad Gestacional , Humanos , Ventrículos Laterales/irrigación sanguínea , Ventrículos Laterales/ultraestructura , Masculino , Microscopía Electrónica de Rastreo , Embarazo , Vénulas/embriología , Vénulas/ultraestructura
11.
Free Radic Biol Med ; 42(10): 1610-23, 2007 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-17448908

RESUMEN

We evaluated the contribution of superoxide anion (O2*-) generated by NADPH oxidase or mitochondria in the rostral ventrolateral medulla (RVLM), where sympathetic premotor neurons for arterial pressure maintenance are located, on cardiovascular depression induced by inducible nitric oxide synthase-derived NO after Escherichia coli lipopolysaccharide (LPS) treatment. In Sprague-Dawley rats maintained under propofol anesthesia, microinjection of LPS bilaterally into the RVLM induced progressive hypotension, bradycardia, and reduction in sympathetic vasomotor outflow over our 240-min observation period. This was accompanied by an increase in O2*- production (60-240 min) in the RVLM, alongside phosphorylation of p47(phox) or p67(phox), upregulation of gp91(phox) or p47(phox) protein, and increase in Rac-1 or NADPH oxidase activity (60-120 min), and a depression of mitochondrial respiratory enzyme activity (120-240 min). Whereas inhibition of NADPH oxidase or knockdown of the gp91(phox) or p47(phox) gene blunted the early phase (60-150 min), coenzyme Q10 or mitochondrial K(ATP) channel inhibitor antagonized the delayed phase (120-240 min) of LPS-induced increase in O2*- production in RVLM and cardiovascular depression. We conclude that, whereas NADPH oxidase-derived O2*- in RVLM participates predominantly in the early phase, O2*- generated by depression in mitochondrial respiratory enzyme activity or opening of mitoK(ATP) channels mediates the delayed phase of LPS-induced cardiovascular depression.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Ventrículos Laterales/enzimología , Mitocondrias/enzimología , NADPH Oxidasas/metabolismo , Superóxidos/metabolismo , Animales , Enfermedades Cardiovasculares/inducido químicamente , Endotoxinas/toxicidad , Ventrículos Laterales/ultraestructura , Masculino , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , NADPH Oxidasa 2 , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/genética , Fosforilación , Canales de Potasio/metabolismo , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba , Proteína de Unión al GTP rac1/metabolismo
12.
Aging Cell ; 5(2): 139-52, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16626393

RESUMEN

In the adult mouse brain, the subventricular zone (SVZ) is a neurogenic stem cell niche only 4-5 cell diameters thick. Within this narrow zone, a unique microenvironment supports stem cell self-renewal, gliogenesis or neurogenesis lineage decisions and tangential migration of newly generated neurons out of the SVZ and into the olfactory bulb. However, with aging, SVZ neurogenesis declines. Here, we examine the dynamic interplay between SVZ cytoarchitecture and neurogenesis through aging. Assembly of high-resolution electron microscopy images of corresponding coronal sections from 2-, 10- and 22-month-old mice into photomontages reveal a thinning of the SVZ with age. Following a 2-h BrdU pulse, we detect a significant decrease in cell proliferation from 2 to 22 months. Neuroblast numbers decrease with age, as do transitory amplifying progenitor cells, while both SVZ astrocytes and adjacent ependymal cells remain relatively constant. At 22 months, only residual pockets of neurogenesis remain and neuroblasts become restricted to the anterior dorsolateral horn of the SVZ. Within this dorsolateral zone many key components of the younger neurogenic niche are maintained; however, in the aged SVZ, increased numbers of SVZ astrocytes are found interposed within the ependyma. These astrocytes co-label with markers to ependymal cells and astrocytes, form intercellular adherens junctions with neighboring ependymal cells, and some possess multiple basal bodies of cilia within their cytoplasm. Together, these data reveal an age-related, progressive restriction of SVZ neurogenesis to the dorsolateral aspect of the lateral ventricle with increased numbers of SVZ astrocytes interpolated within the ependyma.


Asunto(s)
Envejecimiento/fisiología , Ventrículos Laterales/citología , Ventrículos Laterales/fisiología , Neuronas/fisiología , Animales , Apoptosis , Astrocitos/ultraestructura , Proliferación Celular , Epéndimo/citología , Epéndimo/ultraestructura , Ventrículos Laterales/ultraestructura , Ratones , Neuronas/citología , Células Madre/metabolismo
13.
Redox Biol ; 11: 231-239, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28012438

RESUMEN

Fulfilling a bevy of biological roles, copper is an essential metal for healthy brain function. Cu dyshomeostasis has been demonstrated to be involved in some neurological conditions including Menkes and Alzheimer's diseases. We have previously reported localized Cu-rich aggregates in astrocytes of the subventricular zone (SVZ) in rodent brains with Cu concentrations in the hundreds of millimolar. Metallothionein, a cysteine-rich protein critical to metal homeostasis and known to participate in a variety of neuroprotective and neuroregenerative processes, was proposed as a binding protein. Here, we present an analysis of metallothionein(1,2) knockout (MTKO) mice and age-matched controls using X-ray fluorescence microscopy. In large structures such as the corpus callosum, cortex, and striatum, there is no significant difference in Cu, Fe, or Zn concentrations in MTKO mice compared to age-matched controls. In the astrocyte-rich subventricular zone where Cu-rich aggregates reside, approximately 1/3 as many Cu-rich aggregates persist in MTKO mice resulting in a decrease in periventricular Cu concentration. Aggregates in both wild-type and MTKO mice show XANES spectra characteristic of CuxSy multimetallic clusters and have similar [S]/[Cu] ratios. Consistent with assignment as a CuxSy multimetallic cluster, the astrocyte-rich SVZ of both MTKO and wild-type mice exhibit autofluorescent bodies, though MTKO mice exhibit fewer. Furthermore, XRF imaging of Au-labeled lysosomes and ubiquitin demonstrates a lack of co-localization with Cu-rich aggregates suggesting they are not involved in a degradation pathway. Overall, these data suggest that Cu in aggregates is bound by either metallothionein-3 or a yet unknown protein similar to metallothionein.


Asunto(s)
Astrocitos/metabolismo , Cobre/química , Ventrículos Laterales/metabolismo , Metalotioneína/deficiencia , Animales , Astrocitos/ultraestructura , Biomarcadores/metabolismo , Cationes Bivalentes , Corteza Cerebral/metabolismo , Corteza Cerebral/ultraestructura , Precipitación Química , Cuerpo Calloso/metabolismo , Cuerpo Calloso/ultraestructura , Cuerpo Estriado/metabolismo , Cuerpo Estriado/ultraestructura , Eliminación de Gen , Expresión Génica , Hierro/química , Ventrículos Laterales/ultraestructura , Proteínas de Membrana de los Lisosomas/genética , Proteínas de Membrana de los Lisosomas/metabolismo , Lisosomas/metabolismo , Lisosomas/ultraestructura , Metalotioneína/genética , Ratones , Ratones Noqueados , Imagen Óptica , Ratas , Ratas Sprague-Dawley , Espectrometría por Rayos X , Ubiquitina/genética , Ubiquitina/metabolismo , Zinc/química
14.
Transl Stroke Res ; 8(4): 362-373, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28102508

RESUMEN

We previously found that hematoma worsens hydrocephalus after intraventricular hemorrhage (IVH) via increasing iron deposition and aggravating ependymal cilia injury; therefore, promoting hematoma absorption may be a promising strategy for IVH. Recently, some investigations imply that simvastatin has the ability of accelerating hematoma absorption. Thus, this study was designed to examine the efficacy of simvastatin for IVH in rats. Intracerebral hemorrhage with ventricular extension was induced in adult male Sprague-Dawley rats after autologous blood injection. Simvastatin or vehicle was administered orally at 1 day after IVH and then daily for 1 week. MRI studies were performed to measure the volumes of intracranial hematoma and lateral ventricle at days 1, 3, 7, 14, and 28 after IVH. Motor and neurocognitive functions were assessed at days 1 to 7 and 23 to 28, respectively. Iron deposition, iron-related protein expression, ependymal damage, and histology were detected at day 28. Expression of CD36 scavenger receptor (facilitating phagocytosis) was examined at day 3 after IVH using western blotting and immunofluorescence. Simvastatin significantly increased hematoma absorption ratio, reduced ventricular volume, and attenuated neurological dysfunction post-IVH. In addition, less iron accumulation and more cilia survival was observed in the simvastatin group when compared with the control. What's more, higher expression of CD36 was detected around the hematoma after simvastatin administration. Simvastatin significantly enhanced brain hematoma absorption, alleviated hydrocephalus, and improved neurological recovery after experimental IVH, which may in part by upregulating CD36 expression. Our data suggest that early simvastatin use may be a novel therapy for IVH patients.


Asunto(s)
Antígenos CD36/metabolismo , Hematoma/tratamiento farmacológico , Hidrocefalia/tratamiento farmacológico , Hipolipemiantes/uso terapéutico , Simvastatina/uso terapéutico , Regulación hacia Arriba/efectos de los fármacos , Animales , Encéfalo/patología , Encéfalo/ultraestructura , Antígeno CD11b/metabolismo , Recuento de Células , Hemorragia Cerebral/complicaciones , Hemorragia Cerebral/diagnóstico por imagen , Modelos Animales de Enfermedad , Epéndimo/metabolismo , Epéndimo/patología , Epéndimo/ultraestructura , Ferritinas/metabolismo , Ferritinas/ultraestructura , Estudios de Seguimiento , Hematoma/diagnóstico por imagen , Hematoma/etiología , Hidrocefalia/diagnóstico por imagen , Hidrocefalia/etiología , Ventrículos Laterales/diagnóstico por imagen , Ventrículos Laterales/patología , Ventrículos Laterales/ultraestructura , Imagen por Resonancia Magnética , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Microscopía Electrónica de Transmisión , Examen Neurológico , Neuronas/metabolismo , Neuronas/patología , Neuronas/ultraestructura , Ratas , Ratas Sprague-Dawley
16.
Brain Struct Funct ; 221(1): 239-60, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25300351

RESUMEN

New subventricular zone (SVZ)-derived neuroblasts that migrate via the rostral migratory stream are continuously added to the olfactory bulb (OB) of the adult rodent brain. Anosmin-1 (A1) is an extracellular matrix protein that binds to FGF receptor 1 (FGFR1) to exert its biological effects. When mutated as in Kallmann syndrome patients, A1 is associated with severe OB morphogenesis defects leading to anosmia and hypogonadotropic hypogonadism. Here, we show that A1 over-expression in adult mice strongly increases proliferation in the SVZ, mainly with symmetrical divisions, and produces substantial morphological changes in the normal SVZ architecture, where we also report the presence of FGFR1 in almost all SVZ cells. Interestingly, for the first time we show FGFR1 expression in the basal body of primary cilia in neural progenitor cells. Additionally, we have found that A1 over-expression also enhances neuroblast motility, mainly through FGFR1 activity. Together, these changes lead to a selective increase in several GABAergic interneuron populations in different OB layers. These specific alterations in the OB would be sufficient to disrupt the normal processing of sensory information and consequently alter olfactory memory. In summary, this work shows that FGFR1-mediated A1 activity plays a crucial role in the continuous remodelling of the adult OB.


Asunto(s)
Proteínas de la Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/fisiología , Ventrículos Laterales/fisiología , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/fisiología , Neurogénesis , Bulbo Olfatorio/fisiología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Animales , División Celular , Movimiento Celular , Células Cultivadas , Proteínas de la Matriz Extracelular/genética , Humanos , Interneuronas/metabolismo , Interneuronas/fisiología , Ventrículos Laterales/metabolismo , Ventrículos Laterales/ultraestructura , Memoria a Corto Plazo/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Vías Nerviosas/metabolismo , Vías Nerviosas/fisiología , Vías Nerviosas/ultraestructura , Odorantes , Bulbo Olfatorio/metabolismo , Percepción Olfatoria/fisiología
17.
Cell Death Dis ; 7: e2223, 2016 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-27171265

RESUMEN

Current treatments for demyelinating diseases are generally only capable of ameliorating the symptoms, with little to no effect in decreasing myelin loss nor promoting functional recovery. Mesenchymal stem cells (MSCs) have been shown by many researchers to be a potential therapeutic tool in treating various neurodegenerative diseases, including demyelinating disorders. However, in the majority of the cases, the effect was only observed locally, in the area surrounding the graft. Thus, in order to achieve general remyelination in various brain structures simultaneously, bone marrow-derived MSCs were transplanted into the lateral ventricles (LVs) of the cuprizone murine model. In this manner, the cells may secrete soluble factors into the cerebrospinal fluid (CSF) and boost the endogenous oligodendrogenic potential of the subventricular zone (SVZ). As a result, oligodendrocyte progenitor cells (OPCs) were recruited within the corpus callosum (CC) over time, correlating with an increased myelin content. Electrophysiological studies, together with electron microscopy (EM) analysis, indicated that the newly formed myelin correctly enveloped the demyelinated axons and increased signal transduction through the CC. Moreover, increased neural stem progenitor cell (NSPC) proliferation was observed in the SVZ, possibly due to the tropic factors released by the MSCs. In conclusion, the findings of this study revealed that intraventricular injections of MSCs is a feasible method to elicit a paracrine effect in the oligodendrogenic niche of the SVZ, which is prone to respond to the factors secreted into the CSF and therefore promoting oligodendrogenesis and functional remyelination.


Asunto(s)
Enfermedades Desmielinizantes/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Células-Madre Neurales/citología , Oligodendroglía/citología , Comunicación Paracrina/fisiología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Diferenciación Celular , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Cuerpo Calloso/metabolismo , Cuerpo Calloso/ultraestructura , Cuprizona , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/genética , Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Inyecciones Intraventriculares , Péptidos y Proteínas de Señalización Intercelular/líquido cefalorraquídeo , Péptidos y Proteínas de Señalización Intercelular/genética , Ventrículos Laterales/metabolismo , Ventrículos Laterales/ultraestructura , Células Madre Mesenquimatosas/fisiología , Ratones , Vaina de Mielina/metabolismo , Vaina de Mielina/ultraestructura , Células-Madre Neurales/fisiología , Oligodendroglía/fisiología , Transducción de Señal
18.
J Neurosci ; 22(6): 2255-64, 2002 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-11896165

RESUMEN

The subventricular zone (SVZ) is the largest germinal layer in the adult mammalian brain and comprises stem cells, transit-amplifying progenitors, and committed neuroblasts. Although the SVZ contains the highest concentration of dividing cells in the adult brain, the intracellular mechanisms controlling their proliferation have not been elucidated. We show here that loss of the cyclin-dependent kinase inhibitor p27Kip1 has very specific effects on a population of CNS progenitors responsible for adult neurogenesis. Using bromodeoxyuridine and [(3)H]thymidine incorporation to label cells in S phase and cell-specific markers and electron microscopy to identify distinct cell types, we compared the SVZ structure and proliferation characteristics of wild-type and p27Kip1-null mice. Loss of p27Kip1 had no effect on the number of stem cells but selectively increased the number of the transit-amplifying progenitors concomitantly with a reduction in the number of neuroblasts. We conclude that cell-cycle regulation of SVZ adult progenitors is remarkably cell-type specific, with p27Kip1 being a key regulator of the cell division of the transit-amplifying progenitors.


Asunto(s)
Neuronas/citología , Células Madre/citología , Células Madre/fisiología , Proteínas Supresoras de Tumor/deficiencia , Animales , Apoptosis/genética , Bromodesoxiuridina/metabolismo , Recuento de Células , Ciclo Celular , Proteínas de Ciclo Celular/genética , Diferenciación Celular/efectos de los fármacos , División Celular/genética , Células Cultivadas , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Ventrículos Laterales/citología , Ventrículos Laterales/ultraestructura , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/metabolismo , Esferoides Celulares/citología , Células Madre/metabolismo , Timidina/metabolismo , Proteínas Supresoras de Tumor/genética
19.
J Vis Exp ; (100): e52853, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-26067390

RESUMEN

Multiciliated ependymal cells line the ventricles in the adult brain. Abnormal function or structure of ependymal cilia is associated with various neurological deficits. The current ex vivo live imaging of motile ependymal cilia technique allows for a detailed study of ciliary dynamics following several steps. These steps include: mice euthanasia with carbon dioxide according to protocols of The University of Toledo's Institutional Animal Care and Use Committee (IACUC); craniectomy followed by brain removal and sagittal brain dissection with a vibratome or sharp blade to obtain very thin sections through the brain lateral ventricles, where the ependymal cilia can be visualized. Incubation of the brain's slices in a customized glass-bottom plate containing Dulbecco's Modified Eagle's Medium (DMEM)/High-Glucose at 37 °C in the presence of 95%/5% O2/CO2 mixture is essential to keep the tissue alive during the experiment. A video of the cilia beating is then recorded using a high-resolution differential interference contrast microscope. The video is then analyzed frame by frame to calculate the ciliary beating frequency. This allows distinct classification of the ependymal cells into three categories or types based on their ciliary beating frequency and angle. Furthermore, this technique allows the use of high-speed fluorescence imaging analysis to characterize the unique intracellular calcium oscillation properties of ependymal cells as well as the effect of pharmacological agents on the calcium oscillations and the ciliary beating frequency. In addition, this technique is suitable for immunofluorescence imaging for ciliary structure and ciliary protein localization studies. This is particularly important in disease diagnosis and phenotype studies. The main limitation of the technique is attributed to the decrease in live motile cilia movement as the brain tissue starts to die.


Asunto(s)
Cilios/fisiología , Epéndimo/fisiología , Epéndimo/ultraestructura , Ventrículos Laterales/fisiología , Ventrículos Laterales/ultraestructura , Microscopía de Interferencia/métodos , Animales , Ratones , Ratones Endogámicos C57BL
20.
Neurotoxicology ; 50: 46-55, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26215120

RESUMEN

It is still being debated whether neurogenesis in the subventricular zone (SVZ) is enhanced in response to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) injury in the adult mouse brain. Our previous studies provided evidence that MPTP induces apoptosis of migrating neuroblasts (neural progenitor cells, A cells) in the SVZ and rostral migratory stream (RMS). We investigated cellular kinetics in the adult SVZ and olfactory bulb (OB) after MPTP damage. Cells were labeled with bromodeoxyuridine (BrdU), and the effects of MPTP on the survival and fate of migrating and residing neuroblasts were evaluated. Two days after BrdU labeling and MPTP treatment, the number of BrdU-positive cells in the SVZ and OB of MPTP-treated mice was significantly lower than in the SVZ and OB of saline controls. Additionally, fewer BrdU-positive cells migrated to the OB of treated mice than to that of saline controls, and the cells that did migrate diffused radially into the granule cell layer (GCL) when observed at 7, 14, and 28 days. In the OB GCL, the differentiation of BrdU-positive cells into mature neurons significantly attenuated 14 and 28 days after MPTP injury. Moreover, the impaired neurogenesis was followed by a recovery of A cells in the SVZ and OB, suggesting activation of the self-repair process as a result of MPTP-induced depletion of BrdU-positive cells. Our findings clarify the kinetics underlying neurogenesis in MPTP-treated mice and may contribute to the development of an animal model of Parkinson's disease, and the demonstration of cellular kinetics in SVZ may also provide a new insight into assessing neurogenesis in MPTP-treated mouse.


Asunto(s)
1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Ventrículos Laterales/efectos de los fármacos , Intoxicación por MPTP/inducido químicamente , Intoxicación por MPTP/patología , Neurogénesis/efectos de los fármacos , Bulbo Olfatorio/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Bromodesoxiuridina/metabolismo , Proteínas de Unión al Calcio/metabolismo , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Receptores ErbB/metabolismo , Ventrículos Laterales/fisiología , Ventrículos Laterales/ultraestructura , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/metabolismo , Microscopía Electrónica de Transmisión , Proteínas del Tejido Nervioso/metabolismo , Bulbo Olfatorio/fisiología , Bulbo Olfatorio/ultraestructura , Antígeno Nuclear de Célula en Proliferación/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA