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











Intervalo de año de publicación
1.
Cell Death Dis ; 14(2): 84, 2023 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-36746936

RESUMEN

Maintenance of immunological homeostasis between tolerance and autoimmunity is essential for the prevention of human diseases ranging from autoimmune disease to cancer. Accumulating evidence suggests that p53 can mitigate phagocytosis-induced adjuvanticity thereby promoting immunological tolerance following programmed cell death. Here we identify Inhibitor of Apoptosis Stimulating p53 Protein (iASPP), a negative regulator of p53 transcriptional activity, as a regulator of immunological tolerance. iASPP-deficiency promoted lung adenocarcinoma and pancreatic cancer tumorigenesis, while iASPP-deficient mice were less susceptible to autoimmune disease. Immune responses to iASPP-deficient tumors exhibited hallmarks of immunosuppression, including activated regulatory T cells and exhausted CD8+ T cells. Interestingly, iASPP-deficient tumor cells and tumor-infiltrating myeloid cells, CD4+, and γδ T cells expressed elevated levels of PD-1H, a recently identified transcriptional target of p53 that promotes tolerogenic phagocytosis. Identification of an iASPP/p53 axis of immune homeostasis provides a therapeutic opportunity for both autoimmune disease and cancer.


Asunto(s)
Enfermedades Autoinmunes , Neoplasias , Humanos , Ratones , Animales , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Represoras/metabolismo , Proteínas Inhibidoras de la Apoptosis/metabolismo , Linfocitos T CD8-positivos/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias/genética , Enfermedades Autoinmunes/genética , Línea Celular Tumoral
3.
Front Cell Dev Biol ; 8: 573487, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33123536

RESUMEN

Age-related biological alterations in brain function increase the risk of mild cognitive impairment and dementia, a global problem exacerbated by aging populations in developed nations. Limited pharmacological therapies have resulted in attention turning to the promising role of medicinal plants and dietary supplements in the treatment and prevention of dementia. Sugarcane (Saccharum officinarum L.) top, largely considered as a by-product because of its low sugar content, in fact contains the most abundant amounts of antioxidant polyphenols relative to the rest of the plant. Given the numerous epidemiological studies on the effects of polyphenols on cognitive function, in this study, we analyzed polyphenolic constituents of sugarcane top and examined the effect of sugarcane top ethanolic extract (STEE) on a range of central nervous system functions in vitro and in vivo. Orally administrated STEE rescued spatial learning and memory deficit in the senescence-accelerated mouse prone 8 (SAMP8) mice, a non-transgenic strain that spontaneously develops a multisystemic aging phenotype including pathological features of Alzheimer's disease. This could be correlated with an increased number of hippocampal newborn neurons and restoration of cortical monoamine levels in STEE-fed SAMP8 mice. Global genomic analysis by microarray in cerebral cortices showed multiple potential mechanisms for the cognitive improvement. Gene set enrichment analysis (GSEA) revealed biological processes such as neurogenesis, neuron differentiation, and neuron development were significantly enriched in STEE-fed mice brain compared to non-treated SAMP8 mice. Furthermore, STEE treatment significantly regulated genes involved in neurotrophin signaling, glucose metabolism, and neural development in mice brain. Our in vitro results suggest that STEE treatment enhances the metabolic activity of neuronal cells promoting glucose metabolism with significant upregulation of genes, namely PGK1, PGAM1, PKM, and PC. STEE also stimulated proliferation of human neural stem cells (hNSCs), regulated bHLH factor expression and induced neuronal differentiation and astrocytic process lengthening. Altogether, our findings suggest the potential of STEE as a dietary intervention, with promising implications as a novel nutraceutical for cognitive health.

4.
Stem Cell Reports ; 15(3): 789-802, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32763162

RESUMEN

The subventricular zone of the mammalian brain is the major source of adult born neurons. These neuroblasts normally migrate long distances to the olfactory bulbs but can be re-routed to locations of injury and promote neuroregeneration. Mechanistic understanding and pharmacological targets regulating neuroblast migration is sparse. Furthermore, lack of migration assays limits development of pharmaceutical interventions targeting neuroblast recruitment. We therefore developed a physiologically relevant 3D neuroblast spheroid migration assay that permits the investigation of large numbers of interventions. To verify the assay, 1,012 kinase inhibitors were screened for their effects on migration. Several induced significant increases or decreases in migration. MuSK and PIK3CB were selected as putative targets and their knockdown validated increased neuroblast migration. Thus, compounds identified through this assay system could be explored for their potential in augmenting neuroblast recruitment to sites of injury for neuroregeneration, or for decreasing malignant invasion.


Asunto(s)
Bioensayo/métodos , Movimiento Celular , Neuronas/citología , Esferoides Celulares/citología , Animales , Automatización , Movimiento Celular/efectos de los fármacos , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Colorantes Fluorescentes/metabolismo , Técnicas de Silenciamiento del Gen , Procesamiento de Imagen Asistido por Computador , Ventrículos Laterales/citología , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores Colinérgicos/metabolismo , Reproducibilidad de los Resultados , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Imagen de Lapso de Tiempo
5.
Stem Cells ; 38(9): 1149-1158, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32442340

RESUMEN

Postnatal subventricular zone (pSVZ) stem and progenitor cell proliferation is regulated by several developmental signaling pathways such as Wnt/ß-catenin. However, the molecular regulation of Wnt function in the pSVZ is poorly understood. We previously showed that Wnt signaling is upregulated in an SVZ gliomagenesis in vivo model. As well, the pro-inflammatory molecule Galectin-3 (Gal-3) increases Wnt signaling in cancer cells and is expressed in the SVZ. Therefore, we asked if Gal-3 has a similar function on Wnt signaling in the pSVZ. We interrogated Wnt signaling using a signaling reporter as well as immunohistochemistry and showed that Wnt signaling predominates upstream in the pSVZ lineage but is downregulated in migrating neuroblasts. Biochemical analysis of SVZ cells, in vivo and in neurosphere stem/progenitor cells, showed that Gal-3 physically interacts with multiple forms of ß-catenin, which is a major downstream regulator of Wnt signaling. Functional analyses demonstrated, in vitro and in vivo, that Gal-3 knockdown increases Wnt signaling and conversely that Gal-3 OE inhibits Wnt/ß-catenin signaling in the pSVZ. This latter result suggested that Gal-3, which is consistently increased in brain injury, may decrease pSVZ proliferation. We showed that Gal-3 OE decreased proliferation without altering cell cycle re-entry and that it increased p27Kip1, a molecule which induces cell cycle exit. Our data uncover a novel regulator of Wnt signaling in the SVZ, Gal-3, which does so in a manner opposite to cancer.


Asunto(s)
Galectina 3/metabolismo , Ventrículos Laterales/metabolismo , Vía de Señalización Wnt , Animales , Ciclo Celular , Linaje de la Célula , Proliferación Celular , Regulación hacia Abajo , Ratones Endogámicos C57BL , Unión Proteica , Nicho de Células Madre , beta Catenina/metabolismo
6.
Front Cell Dev Biol ; 8: 600575, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33634096

RESUMEN

Much attention has recently been focused on nutraceuticals, with minimal adverse effects, developed for preventing or treating neurological diseases such as Alzheimer's disease (AD). The present study was conducted to investigate the potential effect on neural development and function of the microalgae Aurantiochytrium sp. as a nutraceutical. To test neuroprotection by the ethanol extract of Aurantiochytrium (EEA) and a derivative, the n-Hexane layer of EEA (HEEA), amyloid-ß-stimulated SH-SY5Y cells, was used as an in vitro AD model. We then assessed the potential enhancement of neurogenesis by EEA and HEEA using murine ex vivo neurospheres. We also administered EEA or HEEA to senescence-accelerated mouse-prone 8 (SAMP8) mice, a non-transgenic strain with accelerated aging and AD-like memory loss for evaluation of spatial learning and memory using the Morris water maze test. Finally, we performed immunohistochemical analysis for assessment of neurogenesis in mice administered EEA. Pretreatment of SH-SY5Y cells with EEA or the squalene-rich fraction of EEA, HEEA, ameliorated amyloid-ß-induced cytotoxicity. Interestingly, only EEA-treated cells showed a significant increase in cell metabolism and intracellular adenosine triphosphate production. Moreover, EEA treatment significantly increased the number of neurospheres, whereas HEEA treatment significantly increased the number of ß-III-tubulin+ young neurons and GFAP+ astrocytes. SAMP8 mice were given 50 mg/kg EEA or HEEA orally for 30 days. EEA and HEEA decreased escape latency in the Morris water maze in SAMP8 mice, indicating improved memory. To detect stem cells and newborn neurons, we administered BrdU for 9 days and measured BrdU+ cells in the dentate gyrus, a neurogenic stem cell niche of the hippocampus. In SAMP8 mice, EEA rapidly and significantly increased the number of BrdU+GFAP+ stem cells and their progeny, BrdU+NeuN+ mature neurons. In conclusion, our data in aggregate indicate that EEA and its constituents could be developed into a nutraceutical for promoting brain health and function against several age-related diseases, particularly AD.

7.
Aging (Albany NY) ; 11(2): 401-422, 2019 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-30654329

RESUMEN

Caffeoylquinic acid (CQA) is a natural polyphenol with evidence of antioxidant and neuroprotective effects and prevention of deficits in spatial learning and memory. We studied the cognitive-enhancing effect of 3,4,5-tricaffeoylquinic acid (TCQA) and explored its cellular and molecular mechanism in the senescence-accelerated mouse prone 8 (SAMP8) model of aging and Alzheimer's disease as well as in human neural stem cells (hNSCs). Mice were fed with 5 mg/kg of TCQA for 30 days and were tested in the Morris water maze (MWM). Brain tissues were collected for immunohistochemical detection of bromodeoxyuridine (BrdU) to detect activated stem cells and newborn neurons. TCQA-treated SAMP8 exhibited significantly improved cognitive performance in MWM compared to water-treated SAMP8. TCQA-treated SAMP8 mice also had significantly higher numbers of BrdU+/glial fibrillary acidic protein (GFAP+) and BrdU+/Neuronal nuclei (NeuN+) cells in the dentate gyrus (DG) neurogenic niche compared with untreated SAMP8. In hNSCs, TCQA induced cell cycle arrest at G0/G1, actin cytoskeleton organization, chromatin remodeling, neuronal differentiation, and bone morphogenetic protein signaling. The neurogenesis promoting effect of TCQA in the DG of SAMP8 mice might explain the cognition-enhancing influence of TCQA observed in our study, and our hNSCs in aggregate suggest a therapeutic potential for TCQA in aging-associated diseases.


Asunto(s)
Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Ácido Quínico/análogos & derivados , Animales , Calcio/metabolismo , Línea Celular , Humanos , Masculino , Memoria/fisiología , Ratones , Ratones Endogámicos , Células-Madre Neurales , Neurogénesis/fisiología , Ácido Quínico/farmacología
8.
Prog Neurobiol ; 170: 37-52, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29654835

RESUMEN

The adult subventricular zone (SVZ) stem cell niche has proven vital for discovering neurodevelopmental mechanisms and holds great potential in medicine for neurodegenerative diseases. Yet the SVZ holds a dark side - it can become tumorigenic. Glioblastomas can arise from the SVZ via cancer stem cells (CSCs). Glioblastoma and other brain cancers often have dismal prognoses since they are resistant to treatment. In this review we argue that the SVZ is susceptible to cancer because it contains stem cells, migratory progenitors and unusual inflammation. Theoretically, SVZ stem cells can convert to CSCs more readily than can postmitotic neural cells. Additionally, the robust long-distance migration of SVZ progenitors can be subverted upon tumorigenesis to an infiltrative phenotype. There is evidence that the SVZ, even in health, exhibits chronic low-grade cellular and molecular inflammation. Its inflammatory response to brain injuries and disease differs from that of other brain regions. We hypothesize that the SVZ inflammatory environment can predispose cells to novel mutations and exacerbate cancer phenotypes. This can be studied in animal models in which human mutations related to cancer are knocked into the SVZ to induce tumorigenesis and the CSC immune interactions that precede full-blown cancer. Importantly inflammation can be pharmacologically modulated providing an avenue to brain cancer management and treatment. The SVZ is accessible by virtue of its location surrounding the lateral ventricles and CSCs in the SVZ can be targeted with a variety of pharmacotherapies. Thus, the SVZ can yield aggressive tumors but can be targeted via several strategies.


Asunto(s)
Neoplasias Encefálicas/fisiopatología , Inflamación/fisiopatología , Nicho de Células Madre/inmunología , Animales , Humanos , Ventrículos Laterales
9.
EMBO J ; 37(10)2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29661885

RESUMEN

Many long non-coding RNAs (lncRNAs) are expressed during central nervous system (CNS) development, yet their in vivo roles and mechanisms of action remain poorly understood. Paupar, a CNS-expressed lncRNA, controls neuroblastoma cell growth by binding and modulating the activity of transcriptional regulatory elements in a genome-wide manner. We show here that the Paupar lncRNA directly binds KAP1, an essential epigenetic regulatory protein, and thereby regulates the expression of shared target genes important for proliferation and neuronal differentiation. Paupar promotes KAP1 chromatin occupancy and H3K9me3 deposition at a subset of distal targets, through the formation of a ribonucleoprotein complex containing Paupar, KAP1 and the PAX6 transcription factor. Paupar-KAP1 genome-wide co-occupancy reveals a fourfold enrichment of overlap between Paupar and KAP1 bound sequences, the majority of which also appear to associate with PAX6. Furthermore, both Paupar and Kap1 loss-of-function in vivo disrupt olfactory bulb neurogenesis. These observations provide important conceptual insights into the trans-acting modes of lncRNA-mediated epigenetic regulation and the mechanisms of KAP1 genomic recruitment, and identify Paupar and Kap1 as regulators of neurogenesis in vivo.


Asunto(s)
Cromatina/genética , Células-Madre Neurales/citología , Neuroblastoma/patología , Neurogénesis , Bulbo Olfatorio/citología , ARN Largo no Codificante/metabolismo , Proteína 28 que Contiene Motivos Tripartito/metabolismo , Animales , Animales Recién Nacidos , Ciclo Celular , Proliferación Celular , Células Cultivadas , Epigénesis Genética , Genómica , Ratones , Células-Madre Neurales/metabolismo , Neuroblastoma/genética , Neuroblastoma/metabolismo , Bulbo Olfatorio/metabolismo , Factor de Transcripción PAX6/genética , Factor de Transcripción PAX6/metabolismo , ARN Largo no Codificante/genética , Elementos Reguladores de la Transcripción , Proteína 28 que Contiene Motivos Tripartito/genética
10.
Sci Rep ; 7(1): 7004, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28765636

RESUMEN

Bioprinting is an emerging technique for the fabrication of living tissues that allows cells to be arranged in predetermined three-dimensional (3D) architectures. However, to date, there are limited examples of bioprinted constructs containing multiple cell types patterned at high-resolution. Here we present a low-cost process that employs 3D printing of aqueous droplets containing mammalian cells to produce robust, patterned constructs in oil, which were reproducibly transferred to culture medium. Human embryonic kidney (HEK) cells and ovine mesenchymal stem cells (oMSCs) were printed at tissue-relevant densities (107 cells mL-1) and a high droplet resolution of 1 nL. High-resolution 3D geometries were printed with features of ≤200 µm; these included an arborised cell junction, a diagonal-plane junction and an osteochondral interface. The printed cells showed high viability (90% on average) and HEK cells within the printed structures were shown to proliferate under culture conditions. Significantly, a five-week tissue engineering study demonstrated that printed oMSCs could be differentiated down the chondrogenic lineage to generate cartilage-like structures containing type II collagen.


Asunto(s)
Bioimpresión/métodos , Células Epiteliales/fisiología , Células Madre Mesenquimatosas/fisiología , Técnicas de Cultivo de Órganos/métodos , Impresión Tridimensional , Ingeniería de Tejidos/métodos , Animales , Proliferación Celular , Células Cultivadas , Cabras , Humanos
11.
Cancer Cell ; 30(4): 578-594, 2016 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-27693047

RESUMEN

Isocitrate dehydrogenase 1 mutations drive human gliomagenesis, probably through neomorphic enzyme activity that produces D-2-hydroxyglutarate. To model this disease, we conditionally expressed Idh1R132H in the subventricular zone (SVZ) of the adult mouse brain. The mice developed hydrocephalus and grossly dilated lateral ventricles, with accumulation of 2-hydroxyglutarate and reduced α-ketoglutarate. Stem and transit amplifying/progenitor cell populations were expanded, and proliferation increased. Cells expressing SVZ markers infiltrated surrounding brain regions. SVZ cells also gave rise to proliferative subventricular nodules. DNA methylation was globally increased, while hydroxymethylation was decreased. Mutant SVZ cells overexpressed Wnt, cell-cycle and stem cell genes, and shared an expression signature with human gliomas. Idh1R132H mutation in the major adult neurogenic stem cell niche causes a phenotype resembling gliomagenesis.


Asunto(s)
Neoplasias Encefálicas/enzimología , Glioma/enzimología , Isocitrato Deshidrogenasa/biosíntesis , Ventrículos Laterales/enzimología , Células Madre Neoplásicas/enzimología , Nicho de Células Madre , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Metilación de ADN , Glioma/genética , Glioma/patología , Isocitrato Deshidrogenasa/genética , Ventrículos Laterales/patología , Ratones , Ratones Transgénicos , Mutación , Células Madre Neoplásicas/patología , Transcriptoma
12.
PLoS One ; 11(1): e0146533, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26799938

RESUMEN

Implanted gradient index lenses have extended the reach of standard multiphoton microscopy from the upper layers of the mouse cortex to the lower cortical layers and even subcortical regions. These lenses have the clarity to visualize dynamic activities, such as calcium transients, with subcellular and millisecond resolution and the stability to facilitate repeated imaging over weeks and months. In addition, behavioral tests can be used to correlate performance with observed changes in network function and structure that occur over time. Yet, this raises the questions, does an implanted microlens have an effect on behavioral tests, and if so, what is the extent of the effect? To answer these questions, we compared the performance of three groups of mice in three common behavioral tests. A gradient index lens was implanted in the prefrontal cortex of experimental mice. We compared their performance with mice that had either a cranial window or a sham surgery. Three presurgical and five postsurgical sets of behavioral tests were performed over seven weeks. Behavioral tests included rotarod, foot fault, and Morris water maze. No significant differences were found between the three groups, suggesting that microlens implantation did not affect performance. The results for the current study clear the way for combining behavioral studies with gradient index lens imaging in the prefrontal cortex, and potentially other regions of the mouse brain, to study structural, functional, and behavioral relationships in the brain.


Asunto(s)
Conducta Animal/fisiología , Implantes Experimentales/efectos adversos , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Corteza Prefrontal/cirugía , Prueba de Desempeño de Rotación con Aceleración Constante/métodos , Animales , Escala de Evaluación de la Conducta , Femenino , Proteínas Fluorescentes Verdes/genética , Procesamiento de Imagen Asistido por Computador , Lentes , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Transgénicos , Microscopía de Fluorescencia por Excitación Multifotónica/instrumentación , Neuroimagen/métodos , Desempeño Psicomotor/fisiología
13.
Proc Natl Acad Sci U S A ; 111(27): 9834-9, 2014 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-24958857

RESUMEN

Inflammation and loss of cell polarity play pivotal roles in neurodegeneration and cancer. A central question in both diseases is how the loss of cell polarity is sensed by cell death machinery. Here, we identify apoptosis-stimulating protein of p53 with signature sequences of ankyrin repeat-, SH3 domain-, and proline-rich region-containing protein 2 (ASPP2), a haploinsufficient tumor suppressor, activator of p53, and regulator of cell polarity, as a transcriptional target of signal transducer and activator of transcription 1 (STAT1). LPS induces ASPP2 expression in murine macrophage and microglial cell lines, a human monocyte cell line, and primary human astrocytes in vitro. LPS and IFNs induce ASPP2 transcription through an NF-κB RELA/p65-independent but STAT1-dependent pathway. In an LPS-induced maternal inflammation mouse model, LPS induces nuclear ASPP2 in vivo at the blood-cerebral spinal fluid barrier (the brain's barrier to inflammation), and ASPP2 mediates LPS-induced apoptosis. Consistent with the role of ASPP2 as a gatekeeper to inflammation, ASPP2-deficient brains possess enhanced neuroinflammation. Elevated ASPP2 expression is also observed in mouse models and human neuroinflammatory disease tissue, where ASPP2 was detected in GFAP-expressing reactive astrocytes that coexpress STAT1. Because the ability of ASPP2 to maintain cellular polarity is vital to CNS development, our findings suggest that the identified STAT1/ASPP2 pathway may connect tumor suppression and cell polarity to neuroinflammation.


Asunto(s)
Polaridad Celular , Encefalitis/fisiopatología , Neoplasias Experimentales/prevención & control , Factor de Transcripción STAT1/fisiología , Transcripción Genética/fisiología , Proteínas Supresoras de Tumor/genética , Animales , Apoptosis , Astrocitos/metabolismo , Regulación de la Expresión Génica/fisiología , Humanos , Ratones
14.
Neurobiol Dis ; 63: 155-64, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24269916

RESUMEN

Angiogenesis is thought to decrease stroke size and improve behavioral outcomes and therefore several clinical trials are seeking to augment it. Galectin-3 (Gal-3) expression increases after middle cerebral artery occlusion (MCAO) and has been proposed to limit damage 3days after stroke. We carried out mild MCAO that damages the striatum but spares the cerebral cortex and SVZ. Gal-3 gene deletion prevented vascular endothelial growth factor (VEGF) upregulation after MCAO. This inhibited post-MCAO increases in endothelial proliferation and angiogenesis in the striatum allowing us to uniquely address the function of angiogenesis in this model of stroke. Apoptosis and infarct size were unchanged in Gal-3(-/-) mice 7 and 14 days after MCAO, suggesting that angiogenesis does not affect lesion size. Microglial and astrocyte activation/proliferation after MCAO was similar in wild type and Gal-3(-/-) mice. In addition, openfield activity, motor hemiparesis, proprioception, reflex, tremors and grooming behaviors were essentially identical between WT and Gal-3(-/-) mice at 1, 3, 7, 10 and 14 days after MCAO, suggesting that penumbral angiogenesis has limited impact on behavioral recovery. In addition to angiogenesis, increased adult subventricular zone (SVZ) neurogenesis is thought to provide neuroprotection after stroke in animal models. SVZ neurogenesis and migration to lesion were overall unaffected by the loss of Gal-3, suggesting no compensation for the lack of angiogenesis in Gal-3(-/-) mice. Because angiogenesis and neurogenesis are usually coordinately regulated, identifying their individual effects on stroke has hitherto been difficult. These results show that Gal-3 is necessary for angiogenesis in stroke in a VEGF-dependant manner, but suggest that angiogenesis may be dispensable for post-stroke endogenous repair, therefore drawing into question the clinical utility of augmenting angiogenesis.


Asunto(s)
Inductores de la Angiogénesis/metabolismo , Galectina 3/deficiencia , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/genética , Trastornos Mentales/etiología , Recuperación de la Función/genética , Animales , Encéfalo/metabolismo , Infarto Encefálico/etiología , Infarto Encefálico/patología , Ventrículos Cerebrales/patología , Circulación Cerebrovascular/genética , Modelos Animales de Enfermedad , Proteína Doblecortina , Galectina 3/genética , Regulación de la Expresión Génica/genética , Gliosis/etiología , Infarto de la Arteria Cerebral Media/patología , Masculino , Trastornos Mentales/genética , Ratones , Ratones Noqueados , Neovascularización Patológica , Neurogénesis/genética , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
J Cell Sci ; 124(Pt 14): 2438-47, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21693585

RESUMEN

The adult brain subventricular zone (SVZ) produces neuroblasts that migrate through the rostral migratory stream (RMS) to the olfactory bulb (OB) in a specialized niche. Galectin-3 (Gal-3) regulates proliferation and migration in cancer and is expressed by activated macrophages after brain injury. The function of Gal-3 in the normal brain is unknown, but we serendipitously found that it was expressed by ependymal cells and SVZ astrocytes in uninjured mice. Ependymal cilia establish chemotactic gradients and astrocytes form glial tubes, which combine to aid neuroblast migration. Whole-mount preparations and electron microscopy revealed that both ependymal cilia and SVZ astrocytes were disrupted in Gal3(-/-) mice. Interestingly, far fewer new BrdU(+) neurons were found in the OB of Gal3(-/-) mice, than in wild-type mice 2 weeks after labeling. However, SVZ proliferation and cell death, as well as OB differentiation rates were unaltered. This suggested that decreased migration in vivo was sufficient to decrease the number of new OB neurons. Two-photon time-lapse microscopy in forebrain slices confirmed decreased migration; cells were slower and more exploratory in Gal3(-/-) mice. Gal-3 blocking antibodies decreased migration and dissociated neuroblast cell-cell contacts, whereas recombinant Gal-3 increased migration from explants. Finally, we showed that expression of phosphorylated epidermal growth factor receptor (EGFR) was increased in Gal3(-/-) mice. These results suggest that Gal-3 is important in SVZ neuroblast migration, possibly through an EGFR-based mechanism, and reveals a role for this lectin in the uninjured brain.


Asunto(s)
Movimiento Celular/fisiología , Galectina 3/metabolismo , Ventrículos Laterales/citología , Bulbo Olfatorio/citología , Animales , Diferenciación Celular/fisiología , Galectina 3/deficiencia , Ventrículos Laterales/metabolismo , Ratones , Ratones Transgénicos , Microglía/citología , Microglía/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Bulbo Olfatorio/metabolismo
16.
PLoS One ; 4(12): e8122, 2009 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-19956583

RESUMEN

BACKGROUND: The adult subventricular zone (SVZ) contains stem and progenitor cells that generate neuroblasts throughout life. Although it is well accepted that SVZ neuroblasts are migratory, recent evidence suggests their progenitor cells may also exhibit motility. Since stem and progenitor cells are proliferative and multipotential, if they were also able to move would have important implications for SVZ neurogenesis and its potential for repair. METHODOLOGY/PRINCIPAL FINDINGS: We studied whether SVZ stem and/or progenitor cells are motile in transgenic GFP+ slices with two photon time lapse microscopy and post hoc immunohistochemistry. We found that stem and progenitor cells; mGFAP-GFP+ cells, bright nestin-GFP+ cells and Mash1+ cells were stationary in the SVZ and rostral migratory stream (RMS). In our search for motile progenitor cells, we uncovered a population of motile betaIII-tubulin+ neuroblasts that expressed low levels of epidermal growth factor receptor (EGFr). This was intriguing since EGFr drives proliferation in the SVZ and affects migration in other systems. Thus we examined the potential role of EGFr in modulating SVZ migration. Interestingly, EGFr(low) neuroblasts moved slower and in more tortuous patterns than EGFr-negative neuroblasts. We next questioned whether EGFr stimulation affects SVZ cell migration by imaging Gad65-GFP+ neuroblasts in the presence of transforming growth factor alpha (TGF-alpha), an EGFr-selective agonist. Indeed, acute exposure to TGF-alpha decreased the percentage of motile cells by approximately 40%. CONCLUSIONS/SIGNIFICANCE: In summary, the present study directly shows that SVZ stem and progenitor cells are static, that EGFr is retained on some neuroblasts, and that EGFr stimulation negatively regulates migration. This result suggests an additional role for EGFr signaling in the SVZ.


Asunto(s)
Movimiento Celular , Ventrículos Cerebrales/citología , Receptores ErbB/metabolismo , Neuronas/citología , Células Madre/citología , Animales , Movimiento Celular/efectos de los fármacos , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Factor de Crecimiento Transformador alfa/farmacología
17.
J Neuroinflammation ; 5: 44, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18922161

RESUMEN

BACKGROUND: The periventricular subventricular zone (SVZ) contains stem cells and is an area of active neurogenesis and migration. Since inflammation can reduce neurogenesis, we tested whether Theiler's murine encephalomyelitis virus (TMEV) induces inflammation and reduces neurogenesis in the SVZ. METHODS: We performed immmunohistochemistry for the hematopoietic cell marker CD45 throughout the central nervous system and then examined neuroblasts in the SVZ. RESULTS: CD45+ activation (inflammation) occurred early in the forebrain and preceded cerebellar and spinal cord inflammation. Inflammation in the brain was regionally stochastic except for the SVZ and surrounding periventricular regions where it was remarkably pronounced and consistent. In preclinical mice, SVZ neuroblasts emigrated into inflamed periventricular regions. The number of proliferating phoshpohistone3+ cells and Doublecortin+ (Dcx) SVZ neuroblasts was overall unaffected during the periods of greatest inflammation. However the number of Dcx+ and polysialylated neural cell adhesion molecule (PSA-NCAM+) SVZ neuroblasts decreased only after periventricular inflammation abated. CONCLUSION: Our results suggest that after TMEV infection, the SVZ may mount an attempt at neuronal repair via emigration, a process dampened by decreases in neuroblast numbers.


Asunto(s)
Infecciones por Cardiovirus/inmunología , Encefalitis/inmunología , Células Madre Hematopoyéticas/inmunología , Antígenos Comunes de Leucocito/inmunología , Neurogénesis/inmunología , Theilovirus/inmunología , Animales , Biomarcadores/análisis , Biomarcadores/metabolismo , Infecciones por Cardiovirus/fisiopatología , Linaje de la Célula/inmunología , Movimiento Celular/inmunología , Proliferación Celular , Modelos Animales de Enfermedad , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Encefalitis/fisiopatología , Femenino , Células Madre Hematopoyéticas/virología , Histonas/metabolismo , Ventrículos Laterales , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/fisiopatología , Regeneración Nerviosa/inmunología , Plasticidad Neuronal/inmunología , Neuronas/citología , Neuronas/inmunología , Neuropéptidos/metabolismo , Prosencéfalo/inmunología , Prosencéfalo/patología , Prosencéfalo/fisiopatología
18.
Glia ; 54(4): 329-42, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16862532

RESUMEN

Proliferation decreases in the neurogenic subventricular zone (SVZ) of mice after aspiration lesions of the cerebral cortex. We hypothesized that microglial activation may contribute to this given microglial activation attenuates neurogenesis in the hippocampus. Using CD45, CD11b, IB4, and IL-6 immunohistochemistry (IHC), BrdU IHC, and fluorescent bead tracking of peripheral monocytes into the brain, we compared microglial activation in the SVZ to non-neurogenic forebrain regions. SVZ microglia exhibited greater constitutive activation and proliferation than did microglia in non-neurogenic regions. In contrast to the SVZ, the dentate gyrus (DG) contained relatively few CD45(+) cells. After aspiration cerebral cortex lesions, microglia became activated in the cerebral cortex, corpus callosum, and striatum. SVZ microglial activation did not increase, and similarly, microglia in the DG were less activated after injury than in adjacent non-neurogenic regions. We next showed that SVZ microglia are not categorically refractory to activation, since deep cortical contusion injuries increased SVZ microglial activation. Macrophages migrate into the brain during development, but it is unclear if this is recapitulated after injury. Infiltration of microbead-labeled macrophages into the brain did not change after injury, but resident SVZ microglia were induced to migrate toward the injury. Our data show that both constitutive and postlesion levels of microglial activation differ between neurogenic and non-neurogenic regions.


Asunto(s)
Lesiones Encefálicas/patología , Microglía/fisiología , Prosencéfalo/fisiología , Animales , Antimetabolitos , Bromodesoxiuridina , Proliferación Celular , Corteza Cerebral/lesiones , Ventrículos Cerebrales/citología , Ventrículos Cerebrales/fisiología , Plexo Coroideo/citología , Plexo Coroideo/fisiología , Diagnóstico por Imagen , Colorantes Fluorescentes , Inmunohistoquímica , Macrófagos/fisiología , Masculino , Ratones , Microglía/patología , Microesferas , Prosencéfalo/citología , Prosencéfalo/patología , Fijación del Tejido
19.
Prog Neurobiol ; 74(2): 77-99, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15518954

RESUMEN

The subventricular zones (SVZs) are essential sources of new cells in the developing brain and remnants of these germinal zones persist into adulthood. As these cells have the capacity to replenish neurons and glia that are turning over, many investigators have assessed the SVZ's role in replacing neural cells eliminated by brain injuries. A review of the literature reveals that the progenitors within the SVZs are vulnerable to chemical, radiation and ischemia-induced damage, whereas the neural stem cells are resilient. With moderate insults, the SVZ can recover, but it cannot recover after more severe injury. Thus, the vulnerability of these cells has important ramifications when considering therapeutic interventions for the treatment of brain tumors and for the prospect of recovery after ischemia. The cells of the perinatal and adult SVZ not only have the capacity to replenish their own numbers, but they also have the capacity to replace neurons and glia after ischemic and traumatic brain injuries. Moreover, the mechanisms underlying these regenerative responses are beginning to be revealed. By reviewing, comparing and contrasting the responses of the SVZs to different injuries, our goal is to provide a foundation from which current and future studies on the potential of the SVZs for cell replacement can be evaluated.


Asunto(s)
Lesiones Encefálicas/patología , Lesiones Encefálicas/fisiopatología , Ventrículos Cerebrales/patología , Ventrículos Cerebrales/fisiopatología , Regeneración Nerviosa/fisiología , Recuperación de la Función/fisiología , Células Madre/patología , Animales , Antineoplásicos/efectos de la radiación , Lesiones Encefálicas/embriología , Lesiones Encefálicas/etiología , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Ventrículos Cerebrales/embriología , Humanos , Hipoxia-Isquemia Encefálica/embriología , Hipoxia-Isquemia Encefálica/etiología , Hipoxia-Isquemia Encefálica/patología , Hipoxia-Isquemia Encefálica/fisiopatología , Mamíferos , Traumatismos por Radiación/complicaciones , Traumatismos por Radiación/embriología , Traumatismos por Radiación/patología , Traumatismos por Radiación/fisiopatología
20.
Brain Res ; 996(2): 213-26, 2004 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-14697499

RESUMEN

The subventricular zone (SVZ) generates the largest number of migratory cells in the adult brain. SVZ neuroblasts migrate to the olfactory bulbs (OB) in the adult, whereas during development, SVZ cells migrate into many adjacent nuclei. Previously, we showed that cerebral cortex injury in the adult causes molecular and cellular changes which may recapitulate the developmental migratory directions. Consistent with this, growth factors, as well as models of illness or injury can cause adult SVZ cells to migrate into non-olfactory bulb nuclei. Here, we tested the hypothesis that cerebral cortex injury in the adult mouse induces changes in migration, by labeling adult SVZ cells with a retroviral vector and examining the distribution of cells 4 days and 3 weeks later. Four days after cortical lesions, disproportionately fewer retrovirally-labeled cells had migrated to the olfactory bulb in lesioned mice than in controls. Conversely, the number of cells found in non-olfactory bulb regions (primarily the area of the lesion and the corpus callosum) was increased in lesioned mice. The morphology of these emigrated cells suggested that they were differentiating into glial cells. Three weeks after cortical injury, the majority of retrovirally-labeled cells in both groups of mice had migrated into the granule and periglomerular layers of the olfactory bulb. At 3 weeks, we still observed retrovirally-labeled glial cells in the corpus callosum and in the area of the injury in lesioned mice. These results suggest that cortical lesions cause a transient change in migration patterns of SVZ progeny, which is characterized by decreases in migration to the olfactory bulb but increased migration towards the injury. Our studies also suggest that cortical lesions induce the production of new glial cells which survive for at least 3 weeks after injury. The data support the concept that in the adult, SVZ cells can generate progeny that migrate towards injured areas and thus potentially be harnessed for neural repair.


Asunto(s)
Movimiento Celular/fisiología , Corteza Cerebral/citología , Bulbo Olfatorio/citología , Células Madre/fisiología , Fosfatasa Alcalina/metabolismo , Animales , Diferenciación Celular , Corteza Cerebral/lesiones , Cuerpo Calloso/citología , Cuerpo Calloso/fisiología , Cartilla de ADN , Inmunohistoquímica , Masculino , Ratones , Neuroglía/citología , Plasticidad Neuronal/fisiología , Neuronas/citología , Bulbo Olfatorio/fisiología , Reacción en Cadena de la Polimerasa , Retroviridae , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA