Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 1584, 2024 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-38383565

RESUMEN

Astrocytes, a type of glial cell in the central nervous system (CNS), adopt diverse states in response to injury that are influenced by their location relative to the insult. Here, we describe a platform for spatially resolved, single-cell transcriptomics and proteomics, called tDISCO (tissue-digital microfluidic isolation of single cells for -Omics). We use tDISCO alongside two high-throughput platforms for spatial (Visium) and single-cell transcriptomics (10X Chromium) to examine the heterogeneity of the astrocyte response to a cortical ischemic stroke in male mice. We show that integration of Visium and 10X Chromium datasets infers two astrocyte populations, proximal or distal to the injury site, while tDISCO determines the spatial boundaries and molecular profiles that define these populations. We find that proximal astrocytes show differences in lipid shuttling, with enriched expression of Apoe and Fabp5. Our datasets provide a resource for understanding the roles of astrocytes in stroke and showcase the utility of tDISCO for hypothesis-driven, spatially resolved single-cell experiments.


Asunto(s)
Astrocitos , Accidente Cerebrovascular , Ratones , Animales , Masculino , Astrocitos/metabolismo , Sistema Nervioso Central/metabolismo , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/metabolismo , Perfilación de la Expresión Génica , Cromo/metabolismo
2.
J Proteome Res ; 22(10): 3242-3253, 2023 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-37651704

RESUMEN

Proteome profiles of precious tissue samples have great clinical potential for accelerating disease biomarker discovery and promoting novel strategies for early diagnosis and treatment. However, tiny clinical tissue samples are often difficult to handle and analyze with conventional proteomic methods. Automated digital microfluidic (DMF) workflows facilitate the manipulation of size-limited tissue samples. Here, we report the assessment of a DMF microproteomics workflow enabled by a photocleavable surfactant for proteomic analysis of minute tissue samples. The surfactant 4-hexylphenylazosulfonate (Azo) was found to facilitate fast droplet movement on DMF and enhance the proteomics analysis. Comparisons of Azo and n-Dodecyl ß-d-maltoside (DDM) using small samples of HeLa digest standards and MCF-7 cell digests revealed distinct differences at the peptide level despite similar results at the protein level. The DMF microproteomics workflow was applied for the sample preparation of ∼3 µg biopsies from murine brain tissue. A total of 1969 proteins were identified in three samples, including established neural biomarkers and proteins related to synaptic signaling. Going forward, we propose that the Azo-enabled DMF workflow has the potential to advance the practical clinical application of DMF for the analysis of size-limited tissue samples.

3.
Proc Natl Acad Sci U S A ; 119(49): e2207824119, 2022 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-36454756

RESUMEN

Revealing the molecular events associated with reprogramming different somatic cell types to pluripotency is critical for understanding the characteristics of induced pluripotent stem cell (iPSC) therapeutic derivatives. Inducible reprogramming factor transgenic cells or animals-designated as secondary (2°) reprogramming systems-not only provide excellent experimental tools for such studies but also offer a strategy to study the variances in cellular reprogramming outcomes due to different in vitro and in vivo environments. To make such studies less cumbersome, it is desirable to have a variety of efficient reprogrammable mouse systems to induce successful mass reprogramming in somatic cell types. Here, we report the development of two transgenic mouse lines from which 2° cells reprogram with unprecedented efficiency. These systems were derived by exposing primary reprogramming cells containing doxycycline-inducible Yamanaka factor expression to a transient interruption in transgene expression, resulting in selection for a subset of clones with robust transgene response. These systems also include reporter genes enabling easy readout of endogenous Oct4 activation (GFP), indicative of pluripotency, and reprogramming transgene expression (mCherry). Notably, somatic cells derived from various fetal and adult tissues from these 2° mouse lines gave rise to highly efficient and rapid reprogramming, with transgene-independent iPSC colonies emerging as early as 1 wk after induction. These mouse lines serve as a powerful tool to explore sources of variability in reprogramming and the mechanistic underpinnings of efficient reprogramming systems.


Asunto(s)
Reprogramación Celular , Doxiciclina , Animales , Ratones , Ratones Transgénicos , Reprogramación Celular/genética , Transgenes , Células Clonales , Doxiciclina/farmacología
4.
Front Neurosci ; 16: 917071, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36061596

RESUMEN

Direct neuronal reprogramming, the process whereby a terminally differentiated cell is converted into an induced neuron without traversing a pluripotent state, has tremendous therapeutic potential for a host of neurodegenerative diseases. While there is strong evidence for astrocyte-to-neuron conversion in vitro, in vivo studies in the adult brain are less supportive or controversial. Here, we set out to enhance the efficacy of neuronal conversion of adult astrocytes in vivo by optimizing the neurogenic capacity of a driver transcription factor encoded by the proneural gene Ascl1. Specifically, we mutated six serine phospho-acceptor sites in Ascl1 to alanines (Ascl1 SA 6) to prevent phosphorylation by proline-directed serine/threonine kinases. Native Ascl1 or Ascl1 SA 6 were expressed in adult, murine cortical astrocytes under the control of a glial fibrillary acidic protein (GFAP) promoter using adeno-associated viruses (AAVs). When targeted to the cerebral cortex in vivo, mCherry+ cells transduced with AAV8-GFAP-Ascl1 SA 6-mCherry or AAV8-GFAP-Ascl1-mCherry expressed neuronal markers within 14 days post-transduction, with Ascl1 SA 6 promoting the formation of more mature dendritic arbors compared to Ascl1. However, mCherry expression disappeared by 2-months post-transduction of the AAV8-GFAP-mCherry control-vector. To circumvent reporter issues, AAV-GFAP-iCre (control) and AAV-GFAP-Ascl1 (or Ascl1 SA 6)-iCre constructs were generated and injected into the cerebral cortex of Rosa reporter mice. In all comparisons of AAV capsids (AAV5 and AAV8), GFAP promoters (long and short), and reporter mice (Rosa-zsGreen and Rosa-tdtomato), Ascl1 SA 6 transduced cells more frequently expressed early- (Dcx) and late- (NeuN) neuronal markers. Furthermore, Ascl1 SA 6 repressed the expression of astrocytic markers Sox9 and GFAP more efficiently than Ascl1. Finally, we co-transduced an AAV expressing ChR2-(H134R)-YFP, an optogenetic actuator. After channelrhodopsin photostimulation, we found that Ascl1 SA 6 co-transduced astrocytes exhibited a significantly faster decay of evoked potentials to baseline, a neuronal feature, when compared to iCre control cells. Taken together, our findings support an enhanced neuronal conversion efficiency of Ascl1 SA 6 vs. Ascl1, and position Ascl1 SA 6 as a critical transcription factor for future studies aimed at converting adult brain astrocytes to mature neurons to treat disease.

5.
Front Pharmacol ; 13: 807831, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35847043

RESUMEN

Background: The use of animals and animal-derived products in ethnopharmacological applications is an ancient human practice that continues in many regions today. The local people of the Himalayan region harbor rich traditional knowledge used to treat a variety of human ailments. The present study was intended with the aim of examining animal-based traditional medicine utilized by the population of the Himalayan region of Azad Jammu and Kashmir. Methods: Data were collected from 2017 to 2019 through individual and group interviews. Data on traditional uses of animal products were analyzed, utilizing following indices such as the frequency of citation, use value, relative importance, similarity index, principal component analysis, and cluster analysis to find the highly preferred species in the area. Results: Ethnomedicinal uses of 62 species of vertebrates and invertebrates were documented. Flesh, fat, bone, whole body, milk, skin, egg, head, feathers, bile, blood, and honey were all used in these applications. The uses of 25 animals are reported here for the first time from the study area (mainly insects and birds, including iconic species like the kalij pheasant, Lophura leucomelanos; Himalayan monal, L. impejanus; and western tragopon, Tragopan melanocephalus). The diversity and range of animal-based medicines utilized in these communities are indications of their strong connections with local ecosystems. Conclusion: Our results provide baseline data valuable for the conservation of vertebrate and invertebrate diversity in the region of Himalayan of Azad Jammu and Kashmir. It is possible that screening this fauna for medicinally active chemicals could contribute to the development of new animal-based drugs.

6.
Nat Commun ; 13(1): 2844, 2022 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-35606347

RESUMEN

The cerebral cortex develops from dorsal forebrain neuroepithelial progenitor cells. Following the initial expansion of the progenitor cell pool, these cells generate neurons of all the cortical layers and then astrocytes and oligodendrocytes. Yet, the regulatory pathways that control the expansion and maintenance of the progenitor cell pool are currently unknown. Here we define six basic pathway components that regulate proliferation of cortically specified human neuroepithelial stem cells (cNESCs) in vitro without the loss of cerebral cortex developmental potential. We show that activation of FGF and inhibition of BMP and ACTIVIN A signalling are required for long-term cNESC proliferation. We also demonstrate that cNESCs preserve dorsal telencephalon-specific potential when GSK3, AKT and nuclear CATENIN-ß1 activity are low. Remarkably, regulation of these six pathway components supports the clonal expansion of cNESCs. Moreover, cNESCs differentiate into lower- and upper-layer cortical neurons in vitro and in vivo. The identification of mechanisms that drive the neuroepithelial stem cell self-renewal and differentiation and preserve this potential in vitro is key to developing regenerative and cell-based therapeutic approaches to treat neurological conditions.


Asunto(s)
Glucógeno Sintasa Quinasa 3 , Células Neuroepiteliales , Diferenciación Celular/fisiología , Corteza Cerebral , Humanos , Células Madre , Telencéfalo
7.
Adv Exp Med Biol ; 1212: 31-48, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-30989588

RESUMEN

Direct lineage reprogramming is the conversion of one specialized cell type to another without the need for a pluripotent intermediate. To date, a wide variety of cell types have been successfully generated using direct reprogramming, both in vitro and in vivo. These newly converted cells have the potential to replace cells that are lost to disease and/or injury. In this chapter, we will focus on direct reprogramming in the central nervous system. We will review current progress in the field with regards to all the major neural cell types and explore how cellular heterogeneity, both in the starter cell and target cell population, may have implications for direct reprogramming. Finally, we will discuss new technologies that will improve our understanding of the reprogramming process and aid the development of more specific and efficient future CNS-based reprogramming strategies.


Asunto(s)
Linaje de la Célula , Reprogramación Celular , Sistema Nervioso Central/citología , Neuronas/citología , Diferenciación Celular , Humanos
9.
Cell Stem Cell ; 17(5): 624-34, 2015 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-26456685

RESUMEN

Reactive astrocytes (RAs) have been reported to convert to multipotent neural stem cells (NSCs) capable of neurosphere (NS) formation and multilineage differentiation in vitro. Using genetic tagging, we determined that subventricular zone (SVZ) NSCs give rise to NSs derived from the stroke-injured cortex. We demonstrate that these cells can be isolated from the cortex in two different models of stroke and from different stroke-lesioned cortical regions. Interestingly, SVZ NSCs give rise to a subpopulation of RAs in the cortex that contribute to astrogliosis and scar formation. Last, we show that these SVZ derived RAs can be converted to neurons in vivo by forced expression of Ascl1. Identifying the contribution of cells originating from the SVZ to injury repair has implications for neural regeneration strategies.


Asunto(s)
Astrocitos/citología , Corteza Cerebral/citología , Ventrículos Laterales/citología , Células-Madre Neurales/citología , Accidente Cerebrovascular/patología , Animales , Astrocitos/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Neuronas/patología , Accidente Cerebrovascular/metabolismo
10.
APMIS ; 122(1): 76-80, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23594359

RESUMEN

Alexander disease (AxD) is a neurodegenerative disorder with prominent white matter degeneration and the presence of Rosenthal fibers containing aggregates of glial fibrillary acidic protein (GFAP), and small stress proteins HSP27 and αB-crystallin, and widespread reactive gliosis. AxD is caused by mutations in GFAP, the main astrocyte intermediate filament protein. We previously showed that intermediate filament protein synemin is upregulated in reactive astrocytes after neurotrauma. Here, we examined immunohistochemically the presence of synemin in reactive astrocytes and Rosenthal fibers in two patients with AxD. There was an abundance of GFAP-positive Rosenthal fibers and widespread reactive gliosis in the white matter and subpial regions. Many of the GFAP-positive reactive astrocytes were positive for synemin, and synemin was also present in Rosenthal fibers. We show that synemin is expressed in reactive astrocytes in AxD, and is also present in Rosenthal fibers. The potential role of synemin in AxD pathogenesis remains to be investigated.


Asunto(s)
Enfermedad de Alexander/metabolismo , Astrocitos/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas de Filamentos Intermediarios/metabolismo , Adolescente , Enfermedad de Alexander/genética , Enfermedad de Alexander/patología , Astrocitos/patología , Encéfalo/metabolismo , Encéfalo/patología , Niño , Femenino , Proteína Ácida Fibrilar de la Glía/genética , Gliosis/metabolismo , Gliosis/patología , Humanos , Inmunohistoquímica , Mutación Puntual , Distribución Tisular
11.
Neuroscientist ; 19(6): 567-577, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23797497

RESUMEN

Advances in cellular reprograming have shown that the delivery of specific transcription factors can result in the shift of one cell type to another. Brief forced expression of the four Yamanaka reprogramming factors (Klf4, Sox2, c-Myc, and Oct4) is able to convert many cell types into induced pluripotent stem cells, whereas some lineage specific transcription factors can convert cells from one type directly to another. Numerous strategies have already been developed for deriving neural cell types, with the hopes of better understanding/alleviating neurodegenerative disease. These cells facilitate drug discovery and constitute an autologous source of cells for brain repair, thus, avoiding rejection issues faced by allografts derived from embryonic stem cells. However, proper characterization of the various types of reprogrammed cells and an understanding of how these cells acquire neural fate is necessary before their translation into the clinic. Here, we review the progress, problems, and prospects with reprogrammed cell types with regards to neurodegenerative disease.

12.
Stem Cells ; 30(10): 2320-9, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22887872

RESUMEN

Adult neurogenesis is regulated by a number of cellular players within the neurogenic niche. Astrocytes participate actively in brain development, regulation of the mature central nervous system (CNS), and brain plasticity. They are important regulators of the local environment in adult neurogenic niches through the secretion of diffusible morphogenic factors, such as Wnts. Astrocytes control the neurogenic niche also through membrane-associated factors, however, the identity of these factors and the mechanisms involved are largely unknown. In this study, we sought to determine the mechanisms underlying our earlier finding of increased neuronal differentiation of neural progenitor cells when cocultured with astrocytes lacking glial fibrillary acidic protein (GFAP) and vimentin (GFAP(-/-) Vim(-/-) ). We used primary astrocyte and neurosphere cocultures to demonstrate that astrocytes inhibit neuronal differentiation through a cell-cell contact. GFAP(-/-) Vim(-/-) astrocytes showed reduced endocytosis of Notch ligand Jagged1, reduced Notch signaling, and increased neuronal differentiation of neurosphere cultures. This effect of GFAP(-/-) Vim(-/-) astrocytes was abrogated in the presence of immobilized Jagged1 in a manner dependent on the activity of γ-secretase. Finally, we used GFAP(-/-) Vim(-/-) mice to show that in the absence of GFAP and vimentin, hippocampal neurogenesis under basal conditions as well as after injury is increased. We conclude that astrocytes negatively regulate neurogenesis through the Notch pathway, and endocytosis of Notch ligand Jagged1 in astrocytes and Notch signaling from astrocytes to neural stem/progenitor cells depends on the intermediate filament proteins GFAP and vimentin.


Asunto(s)
Astrocitos/metabolismo , Proteínas de Unión al Calcio/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Neurogénesis/genética , Receptores Notch/genética , Vimentina/genética , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Astrocitos/citología , Proteínas de Unión al Calcio/metabolismo , Comunicación Celular/genética , Diferenciación Celular , Técnicas de Cocultivo , Endocitosis , Regulación del Desarrollo de la Expresión Génica , Proteína Ácida Fibrilar de la Glía , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteína Jagged-1 , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/deficiencia , Cultivo Primario de Células , Receptores Notch/metabolismo , Proteínas Serrate-Jagged , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo , Vimentina/deficiencia , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
13.
J Neuroinflammation ; 9: 144, 2012 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-22734718

RESUMEN

BACKGROUND: In immune-mediated diseases of the central nervous system, astrocytes exposed to interferon-γ (IFN-γ) can express major histocompatibility complex (MHC) class II molecules and antigens on their surface. MHC class II molecules are thought to be delivered to the cell surface by membrane-bound vesicles. However, the characteristics and dynamics of this vesicular traffic are unclear, particularly in reactive astrocytes, which overexpress intermediate filament (IF) proteins that may affect trafficking. The aim of this study was to determine the mobility of MHC class II vesicles in wild-type (WT) astrocytes and in astrocytes devoid of IFs. METHODS: The identity of MHC class II compartments in WT and IF-deficient astrocytes 48 h after IFN-γ activation was determined immunocytochemically by using confocal microscopy. Time-lapse confocal imaging and Alexa Fluor546-dextran labeling of late endosomes/lysosomes in IFN-γ treated cells was used to characterize the motion of MHC class II vesicles. The mobility of vesicles was analyzed using ParticleTR software. RESULTS: Confocal imaging of primary cultures of WT and IF-deficient astrocytes revealed IFN-γ induced MHC class II expression in late endosomes/lysosomes, which were specifically labeled with Alexa Fluor546-conjugated dextran. Live imaging revealed faster movement of dextran-positive vesicles in IFN-γ-treated than in untreated astrocytes. Vesicle mobility was lower in IFN-γ-treated IF-deficient astrocytes than in WT astrocytes. Thus, the IFN-γ-induced increase in the mobility of MHC class II compartments is IF-dependent. CONCLUSIONS: Since reactivity of astrocytes is a hallmark of many CNS pathologies, it is likely that the up-regulation of IFs under such conditions allows a faster and therefore a more efficient delivery of MHC class II molecules to the cell surface. In vivo, such regulatory mechanisms may enable antigen-presenting reactive astrocytes to respond rapidly and in a controlled manner to CNS inflammation.


Asunto(s)
Astrocitos/metabolismo , Compartimento Celular/fisiología , Antígenos de Histocompatibilidad Clase II/metabolismo , Interferón gamma/fisiología , Proteínas de Filamentos Intermediarios/fisiología , Animales , Células Cultivadas , Interferón gamma/genética , Proteínas de Filamentos Intermediarios/genética , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Transporte de Proteínas/genética , Transporte de Proteínas/fisiología , Regulación hacia Arriba/genética , Regulación hacia Arriba/fisiología
14.
Nucleic Acids Res ; 39(4): e24, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21112872

RESUMEN

Single-cell gene expression levels show substantial variations among cells in seemingly homogenous populations. Astrocytes perform many control and regulatory functions in the central nervous system. In contrast to neurons, we have limited knowledge about functional diversity of astrocytes and its molecular basis. To study astrocyte heterogeneity and stem/progenitor cell properties of astrocytes, we used single-cell gene expression profiling in primary mouse astrocytes and dissociated mouse neurosphere cells. The transcript number variability for astrocytes showed lognormal features and revealed that cells in primary cultures to a large extent co-express markers of astrocytes and neural stem/progenitor cells. We show how subpopulations of cells can be identified at single-cell level using unsupervised algorithms and that gene correlations can be used to identify differences in activity of important transcriptional pathways. We identified two subpopulations of astrocytes with distinct gene expression profiles. One had an expression profile very similar to that of neurosphere cells, whereas the other showed characteristics of activated astrocytes in vivo.


Asunto(s)
Astrocitos/clasificación , Perfilación de la Expresión Génica/métodos , Análisis de la Célula Individual , Animales , Astrocitos/metabolismo , Células Cultivadas , Expresión Génica , Ratones , ARN Mensajero/análisis , Células Madre/metabolismo , Regulación hacia Arriba
15.
PLoS One ; 5(4): e10397, 2010 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-20442854

RESUMEN

BACKGROUND: Astroglial cells are activated following injury and up-regulate the expression of the intermediate filament proteins glial fibrillary acidic protein (GFAP) and vimentin. Adult mice lacking the intermediate filament proteins GFAP and vimentin (GFAP(-/-)Vim(-/-)) show attenuated reactive gliosis, reduced glial scar formation and improved regeneration of neuronal synapses after neurotrauma. GFAP(-/-)Vim(-/-) mice exhibit larger brain infarcts after middle cerebral artery occlusion suggesting protective role of reactive gliosis after adult focal brain ischemia. However, the role of astrocyte activation and reactive gliosis in the injured developing brain is unknown. METHODOLOGY/PRINCIPAL FINDINGS: We subjected GFAP(-/-)Vim(-/-) and wild-type mice to unilateral hypoxia-ischemia (HI) at postnatal day 9 (P9). Bromodeoxyuridine (BrdU; 25 mg/kg) was injected intraperitoneally twice daily from P9 to P12. On P12 and P31, the animals were perfused intracardially. Immunohistochemistry with MAP-2, BrdU, NeuN, and S100 antibodies was performed on coronal sections. We found no difference in the hemisphere or infarct volume between GFAP(-/-)Vim(-/-) and wild-type mice at P12 and P31, i.e. 3 and 22 days after HI. At P31, the number of NeuN(+) neurons in the ischemic and contralateral hemisphere was comparable between GFAP(-/-)Vim(-/-) and wild-type mice. In wild-type mice, the number of S100(+) astrocytes was lower in the ipsilateral compared to contralateral hemisphere (65.0+/-50.1 vs. 85.6+/-34.0, p<0.05). In the GFAP(-/-)Vim(-/-) mice, the number of S100(+) astrocytes did not differ between the ischemic and contralateral hemisphere at P31. At P31, GFAP(-/-)Vim(-/-) mice showed an increase in NeuN(+)BrdU(+) (surviving newly born) neurons in the ischemic cortex compared to wild-type mice (6.7+/-7.7; n = 29 versus 2.9+/-3.6; n = 28, respectively, p<0.05), but a comparable number of S100(+)BrdU(+) (surviving newly born) astrocytes. CONCLUSIONS/SIGNIFICANCE: Our results suggest that attenuation of reactive gliosis in the developing brain does not affect the hemisphere or infarct volume after HI, but increases the number of surviving newborn neurons.


Asunto(s)
Infarto Encefálico/patología , Gliosis/patología , Hipoxia-Isquemia Encefálica/patología , Animales , Animales Recién Nacidos , Astrocitos , Supervivencia Celular , Proteína Ácida Fibrilar de la Glía/deficiencia , Ratones , Ratones Noqueados , Neuronas
16.
Mol Cell Neurosci ; 38(2): 170-82, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18434192

RESUMEN

Mammalian SVZ progenitors continuously generate new neurons in the olfactory bulb. After injury, changes in SVZ cell number suggest injury-induced migration. Studies that trace the migration of SVZ precursors into neurodegenerating areas are lacking. Previously, we showed a decrease in BrdU+SVZ cells following excitotoxic damage to the immature rat cortex. Here, we demonstrate that NMDA-induced injury forces endogenous Cell Tracker Green (CTG) labeled VZ/SVZ precursors out of the SVZ into the neurodegenerating cortex. CTG+/Nestin+/Filamin A+ precursors are closely associated with vimentin+/GFAP+/GLAST+ filaments and express both chemokine receptor CXCR4 and Robo1. In the cortex, SVZ-derived progenitors show a progressive expression of developing, migrating and mature neurons and glial markers. CTG+/GFAP+ astrocytes greatly outnumber CTG+/MAP2+/NeuN+ neurons. SVZ-derived progenitors differentiate into both tbr1+ cortical glutamatergic neurons and calretinin+ interneurons. But, there is little integration of these neurons into the existing circuitry, as seen by Fluorogold retrograde tracing from the internal capsule.


Asunto(s)
Movimiento Celular/fisiología , Corteza Motora/patología , Degeneración Nerviosa/patología , Corteza Somatosensorial/patología , Células Madre/citología , Factores de Edad , Animales , Astrocitos/citología , Biomarcadores/metabolismo , Calbindina 2 , Diferenciación Celular/fisiología , División Celular/fisiología , Agonistas de Aminoácidos Excitadores/toxicidad , Femenino , Gliosis/patología , Cápsula Interna/citología , Cápsula Interna/crecimiento & desarrollo , Masculino , Corteza Motora/fisiología , N-Metilaspartato/toxicidad , Degeneración Nerviosa/fisiopatología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Neurotoxinas/toxicidad , Ratas , Ratas Long-Evans , Receptores CXCR4/metabolismo , Receptores Inmunológicos/metabolismo , Proteína G de Unión al Calcio S100/metabolismo , Corteza Somatosensorial/fisiología , Células Madre/metabolismo
17.
J Neurosci Res ; 85(16): 3545-56, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17668855

RESUMEN

Although cleaved caspase-3 is known to be involved in apoptotic cell death mechanisms in neurons, it can also be involved in a nonapoptotic role in astrocytes after postnatal excitotoxic injury. Here we evaluate participation of upstream pathways activating caspase-3 in neurons and glial cells, by studying the intrinsic pathway via caspase-9, the extrinsic pathway via caspase-8, and activation of the p53-dependent pathway. N-methyl-D-aspartate (NMDA) was injected intracortically in 9-day-old postnatal rats, which were sacrificed at several survival times between 4 hr postlesion (pl) and 7 days pl. We analyzed temporal and spatial expression of caspase-8, caspase-9, and p53 and correlation with neuronal and glial markers and caspase-3 activation. Caspase-9 was significantly activated at 10 hpl, strongly correlating with caspase-3. It was present mainly in damaged cortical and hippocampal neurons but was also seen in astrocytes and oligodendrocytes in layer VI and corpus callosum (cc). Caspase-8 showed a diminished correlation with caspase-3. It was present in cortical neurons at 10-72 hpl, showing layer specificity, and also in astroglial and microglial nuclei, mainly in layer VI and cc. p53 Expression increased at 10-72 hpl but did not correlate with caspase-3. p53 Was seen in neurons of the degenerating cortex and in some astrocytes and microglial cells of layer VI and cc. In conclusion, after neonatal excitotoxicity, mainly the mitochondrial intrinsic pathway mediates neuronal caspase-3 and cell death. In astrocytes, caspase-3 is not widely correlated with caspase-8, caspase-9, or p53, except in layer VI-cc astrocytes, where activation of upstream cascades occurs.


Asunto(s)
Encéfalo/enzimología , Encéfalo/crecimiento & desarrollo , Caspasas/metabolismo , Degeneración Nerviosa/enzimología , Neuroglía/enzimología , Neuronas/enzimología , Animales , Apoptosis/fisiología , Encéfalo/fisiopatología , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Caspasa 9/metabolismo , Activación Enzimática/fisiología , Femenino , Gliosis/metabolismo , Masculino , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/fisiopatología , Neurotoxinas/farmacología , Ratas , Ratas Long-Evans , Transducción de Señal/fisiología , Factores de Tiempo , Proteína p53 Supresora de Tumor/metabolismo
18.
Glia ; 55(9): 954-65, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17487878

RESUMEN

Caspase-3 has classically been defined as the main executioner of programmed cell death. However, recent data supports the participation of this protease in non-apoptotic cellular events including cell proliferation, cell cycle regulation, and cellular differentiation. In this study, astroglial cleavage of caspase-3 was analyzed following excitotoxic damage in postnatal rats to determine if its presence is associated with apoptotic cell death, cell proliferation, or cytoskeletal remodeling. A well-characterized in vivo model of excitotoxicity was studied, where damage was induced by intracortical injection of N-methyl-D-asparate (NMDA) in postnatal day 9 rats. Our results demonstrate that cleaved caspase-3 was mainly observed in the nucleus of activated astrocytes in the lesioned hemisphere as early as 4 h postlesion and persisted until the glial scar was formed at 7-14 days, and it was not associated with TUNEL labeling. Caspase-3 enzymatic activity was detected at 10 h and 1 day postlesion in astrocytes, and co-localized with caspase-cleaved fragments of glial fibrillary acidic protein (CCP-GFAP). However, at longer survival times, when astroglial hypertrophy was observed, astroglial caspase-3 did not generally correlate with GFAP cleavage, but instead was associated with de novo expression of vimentin. Moreover, astroglial caspase-3 cleavage was not associated with BrdU incorporation. These results provide further evidence for a nontraditional role of caspases in cellular function that is independent of cell death and suggest that caspase activation is important for astroglial cytoskeleton remodeling following cellular injury.


Asunto(s)
Astrocitos/enzimología , Daño Encefálico Crónico/enzimología , Encéfalo/enzimología , Caspasa 3/metabolismo , Citoesqueleto/enzimología , Gliosis/inducido químicamente , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Astrocitos/patología , Encéfalo/patología , Encéfalo/fisiopatología , Daño Encefálico Crónico/fisiopatología , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Proliferación Celular/efectos de los fármacos , Tamaño de la Célula/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Citoesqueleto/patología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Gliosis/enzimología , Gliosis/fisiopatología , Filamentos Intermedios/efectos de los fármacos , Filamentos Intermedios/metabolismo , Filamentos Intermedios/patología , Masculino , N-Metilaspartato/metabolismo , N-Metilaspartato/toxicidad , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/enzimología , Degeneración Nerviosa/fisiopatología , Neurotoxinas/metabolismo , Neurotoxinas/toxicidad , Ratas , Ratas Long-Evans , Vimentina/metabolismo
19.
Neurosci Lett ; 401(1-2): 71-6, 2006 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-16567040

RESUMEN

Precursor cells have been shown to be affected by oxidative stress, in vivo and vitro, but little is known about the expression of antioxidant mechanisms in neuronal/glial differentiation. We have characterized the expression of Cu/Zn superoxide dismutase (Cu/Zn SOD), one of the main antioxidant proteins involved in the breakdown of superoxide, in the immature rat dorsolateral subventricular zone (SVZ), rostral migratory stream (RMS) and hippocampal subgranular zone (SGZ). Progenitor cells were identified immunohistochemically on cryostat sections by 5'Bromodeoxyuridine (BrdU) incorporation and expressing cells were further characterized using double labeling for progenitor markers. In the SVZ, only a subpopulation of BrdU+ cells, mostly found in the medial SVZ, expressed Cu/Zn SOD. These cells were mostly nestin+ and some were also vimentin+. In contrast, in the lateral SVZ few Cu/Zn SOD+/BrdU+ cells were found. These were primarily nestin+, vimentin-, showed some PSA-NCAM expression, but only a few were NG2+. In the RMS and SGZ virtually all BrdU+ progenitors were Cu/Zn SOD+ and expressed nestin and vimentin. Some RMS cells were also PSA-NCAM+. These findings show a heterogeneous expression of Cu/Zn SOD in restricted cell types in the germinative zones and suggest a role for antioxidant Cu/Zn SOD in progenitor cells of the immature rat brain.


Asunto(s)
Antioxidantes/metabolismo , Encéfalo/crecimiento & desarrollo , Neuroglía/enzimología , Neuronas/enzimología , Células Madre/enzimología , Superóxido Dismutasa/metabolismo , Animales , Animales Recién Nacidos , Biomarcadores/metabolismo , Encéfalo/citología , Encéfalo/enzimología , Bromodesoxiuridina , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Proliferación Celular , Proteínas de Filamentos Intermediarios/metabolismo , Ventrículos Laterales/citología , Ventrículos Laterales/crecimiento & desarrollo , Ventrículos Laterales/fisiología , Proteínas del Tejido Nervioso/metabolismo , Nestina , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Neuroglía/citología , Neuronas/citología , Estrés Oxidativo/fisiología , Ratas , Ratas Long-Evans , Ácidos Siálicos/metabolismo , Vimentina/metabolismo
20.
J Neuroinflammation ; 2(1): 12, 2005 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15929797

RESUMEN

BACKGROUND: In the nervous system, as in other organs, Cu/Zn superoxide dismutase (Cu/Zn SOD) is a key antioxidant enzyme involved in superoxide detoxification in normal cellular metabolism and after cell injury. Although it has been suggested that immature brain has a different susceptibility to oxidative damage than adult brain, the distribution and cell-specific expression of this enzyme in immature brain and after postnatal brain damage has not been documented. METHODS: In this study, we used immunohistochemistry and western blot to analyze the expression of Cu/Zn SOD in intact immature rat brain and in immature rat brain after an NMDA-induced excitotoxic cortical injury performed at postnatal day 9. Double immunofluorescence labelling was used to identify Cu/Zn SOD-expressing cell populations. RESULTS: In intact immature brain, Cu/Zn SOD enzyme was widely expressed at high levels in neurons mainly located in cortical layers II, III and V, in the sub-plate, in the pyriform cortex, in the hippocampus, and in the hypothalamus. Glial fibrillary acidic protein-positive cells only showed Cu/Zn SOD expression in the glia limitans and in scattered cells of the ventricle walls. No expression was detected in interfascicular oligodendroglia, microglia or endothelial cells. Following excitotoxic damage, neuronal Cu/Zn SOD was rapidly downregulated (over 2-4 hours) at the injection site before neurodegeneration signals and TUNEL staining were observed. Later, from 1 day post-lesion onward, an upregulation of Cu/Zn SOD was found due to increased expression in astroglia. A further increase was observed at 3, 5 and 7 days that corresponded to extensive induction of Cu/Zn SOD in highly reactive astrocytes and in the astroglial scar. CONCLUSION: We show here that, in the intact immature brain, the expression of Cu/Zn SOD was mainly found in neurons. When damage occurs, a strong and very rapid downregulation of this enzyme precedes neuronal degeneration, and is followed by an upregulation of Cu/Zn SOD in astroglial cells.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...