Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Cell ; 184(3): 709-722.e13, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33482084

RESUMEN

Neural stem cells (NSCs) in the adult brain transit from the quiescent state to proliferation to produce new neurons. The mechanisms regulating this transition in freely behaving animals are, however, poorly understood. We customized in vivo imaging protocols to follow NSCs for several days up to months, observing their activation kinetics in freely behaving mice. Strikingly, NSC division is more frequent during daylight and is inhibited by darkness-induced melatonin signaling. The inhibition of melatonin receptors affected intracellular Ca2+ dynamics and promoted NSC activation. We further discovered a Ca2+ signature of quiescent versus activated NSCs and showed that several microenvironmental signals converge on intracellular Ca2+ pathways to regulate NSC quiescence and activation. In vivo NSC-specific optogenetic modulation of Ca2+ fluxes to mimic quiescent-state-like Ca2+ dynamics in freely behaving mice blocked NSC activation and maintained their quiescence, pointing to the regulatory mechanisms mediating NSC activation in freely behaving animals.


Asunto(s)
Células Madre Adultas/metabolismo , Calcio/metabolismo , Ritmo Circadiano , Espacio Intracelular/metabolismo , Células-Madre Neurales/metabolismo , Células Madre Adultas/citología , Células Madre Adultas/efectos de los fármacos , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Conducta Animal/efectos de los fármacos , División Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ritmo Circadiano/efectos de los fármacos , Citosol/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Melatonina/metabolismo , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Optogenética , Transducción de Señal/efectos de los fármacos , Triptaminas/farmacología
2.
Cell ; 166(4): 867-880, 2016 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-27518562

RESUMEN

We report that astrocytic insulin signaling co-regulates hypothalamic glucose sensing and systemic glucose metabolism. Postnatal ablation of insulin receptors (IRs) in glial fibrillary acidic protein (GFAP)-expressing cells affects hypothalamic astrocyte morphology, mitochondrial function, and circuit connectivity. Accordingly, astrocytic IR ablation reduces glucose-induced activation of hypothalamic pro-opio-melanocortin (POMC) neurons and impairs physiological responses to changes in glucose availability. Hypothalamus-specific knockout of astrocytic IRs, as well as postnatal ablation by targeting glutamate aspartate transporter (GLAST)-expressing cells, replicates such alterations. A normal response to altering directly CNS glucose levels in mice lacking astrocytic IRs indicates a role in glucose transport across the blood-brain barrier (BBB). This was confirmed in vivo in GFAP-IR KO mice by using positron emission tomography and glucose monitoring in cerebral spinal fluid. We conclude that insulin signaling in hypothalamic astrocytes co-controls CNS glucose sensing and systemic glucose metabolism via regulation of glucose uptake across the BBB.


Asunto(s)
Astrocitos/metabolismo , Glucosa/metabolismo , Hipotálamo/metabolismo , Insulina/metabolismo , Transducción de Señal , Sistema de Transporte de Aminoácidos X-AG/genética , Sistema de Transporte de Aminoácidos X-AG/metabolismo , Animales , Barrera Hematoencefálica , Retículo Endoplásmico/metabolismo , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Homeostasis , Ratones , Mitocondrias/metabolismo , Neuronas/citología , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo
3.
Physiol Rev ; 101(3): 1309-1370, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33000986

RESUMEN

Posttranscriptional gene expression including splicing, RNA transport, translation, and RNA decay provides an important regulatory layer in many if not all molecular pathways. Research in the last decades has positioned RNA-binding proteins (RBPs) right in the center of posttranscriptional gene regulation. Here, we propose interdependent networks of RBPs to regulate complex pathways within the central nervous system (CNS). These are involved in multiple aspects of neuronal development and functioning, including higher cognition. Therefore, it is not sufficient to unravel the individual contribution of a single RBP and its consequences but rather to study and understand the tight interplay between different RBPs. In this review, we summarize recent findings in the field of RBP biology and discuss the complex interplay between different RBPs. Second, we emphasize the underlying dynamics within an RBP network and how this might regulate key processes such as neurogenesis, synaptic transmission, and synaptic plasticity. Importantly, we envision that dysfunction of specific RBPs could lead to perturbation within the RBP network. This would have direct and indirect (compensatory) effects in mRNA binding and translational control leading to global changes in cellular expression programs in general and in synaptic plasticity in particular. Therefore, we focus on RBP dysfunction and how this might cause neuropsychiatric and neurodegenerative disorders. Based on recent findings, we propose that alterations in the entire regulatory RBP network might account for phenotypic dysfunctions observed in complex diseases including neurodegeneration, epilepsy, and autism spectrum disorders.


Asunto(s)
Encefalopatías/metabolismo , Encéfalo/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Humanos
4.
Mol Syst Biol ; 20(4): 321-337, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38365956

RESUMEN

Adult stem cells are important for tissue turnover and regeneration. However, in most adult systems it remains elusive how stem cells assume different functional states and support spatially patterned tissue architecture. Here, we dissected the diversity of neural stem cells in the adult zebrafish brain, an organ that is characterized by pronounced zonation and high regenerative capacity. We combined single-cell transcriptomics of dissected brain regions with massively parallel lineage tracing and in vivo RNA metabolic labeling to analyze the regulation of neural stem cells in space and time. We detected a large diversity of neural stem cells, with some subtypes being restricted to a single brain region, while others were found globally across the brain. Global stem cell states are linked to neurogenic differentiation, with different states being involved in proliferative and non-proliferative differentiation. Our work reveals principles of adult stem cell organization and establishes a resource for the functional manipulation of neural stem cell subtypes.


Asunto(s)
Células Madre Adultas , Células-Madre Neurales , Animales , Pez Cebra/fisiología , Células-Madre Neurales/metabolismo , Neurogénesis , Encéfalo , Diferenciación Celular
5.
EMBO J ; 38(17): e100481, 2019 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-31304985

RESUMEN

Regulation of adult neural stem cell (NSC) number is critical for lifelong neurogenesis. Here, we identified a post-transcriptional control mechanism, centered around the microRNA 204 (miR-204), to control the maintenance of quiescent (q)NSCs. miR-204 regulates a spectrum of transcripts involved in cell cycle regulation, neuronal migration, and differentiation in qNSCs. Importantly, inhibition of miR-204 function reduced the number of qNSCs in the subependymal zone (SEZ) by inducing pre-mature activation and differentiation of NSCs without changing their neurogenic potential. Strikingly, we identified the choroid plexus of the mouse lateral ventricle as the major source of miR-204 that is released into the cerebrospinal fluid to control number of NSCs within the SEZ. Taken together, our results describe a novel mechanism to maintain adult somatic stem cells by a niche-specific miRNA repressing activation and differentiation of stem cells.


Asunto(s)
Plexo Coroideo/química , MicroARNs/genética , Células-Madre Neurales/citología , Adulto , Animales , Ciclo Celular , Diferenciación Celular , Movimiento Celular , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , MicroARNs/líquido cefalorraquídeo , Persona de Mediana Edad , Células-Madre Neurales/química , Nicho de Células Madre
6.
Development ; 142(21): 3661-74, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26395491

RESUMEN

Multiciliated cells are abundant in the epithelial surface of different tissues, including cells lining the walls of the lateral ventricles in the brain and the airway epithelium. Their main role is to control fluid flow and defects in their differentiation are implicated in many human disorders, such as hydrocephalus, accompanied by defects in adult neurogenesis and mucociliary disorder in the airway system. Here we show that Mcidas, which is mutated in human mucociliary clearance disorder, and GemC1 (Gmnc or Lynkeas), previously implicated in cell cycle progression, are key regulators of multiciliated ependymal cell generation in the mouse brain. Overexpression and knockdown experiments show that Mcidas and GemC1 are sufficient and necessary for cell fate commitment and differentiation of radial glial cells to multiciliated ependymal cells. Furthermore, we show that GemC1 and Mcidas operate in hierarchical order, upstream of Foxj1 and c-Myb transcription factors, which are known regulators of ependymal cell generation, and that Notch signaling inhibits GemC1 and Mcidas function. Our results suggest that Mcidas and GemC1 are key players in the generation of multiciliated ependymal cells of the adult neurogenic niche.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Epéndimo/citología , Células Ependimogliales/citología , Células Ependimogliales/metabolismo , Neurogénesis , Proteínas Nucleares/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas de Ciclo Celular/genética , Epéndimo/metabolismo , Factores de Transcripción Forkhead/metabolismo , Ratones , Proteínas Nucleares/genética , Proteínas Proto-Oncogénicas c-myb/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo
7.
Cereb Cortex ; 27(8): 4213-4228, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28472290

RESUMEN

Aging leads to adverse outcomes after traumatic brain injury. The mechanisms underlying these defects, however, are not yet clear. In this study, we found that astrocytes in the aged post-traumatic cerebral cortex develop a significantly reduced proliferative response, resulting in reduced astrocyte numbers in the penumbra. Moreover, experiments of reactive astrocytes in vitro reveal that their diminished proliferation is due to an age-related switch in the division mode with reduced cell-cycle re-entry rather than changes in cell-cycle length. Notably, reactive astrocytes in vivo and in vitro become refractory to stimuli increasing their proliferation during aging, such as Sonic hedgehog signaling. These data demonstrate for the first time that age-dependent, most likely intrinsic changes in the proliferative program of reactive astrocytes result in their severely hampered proliferative response to traumatic injury thereby affecting astrocyte homeostasis.


Asunto(s)
Envejecimiento/fisiología , Astrocitos/fisiología , Lesiones Encefálicas/fisiopatología , Proliferación Celular/fisiología , Homeostasis/fisiología , Corteza Somatosensorial/fisiopatología , Envejecimiento/patología , Animales , Astrocitos/patología , Lesiones Encefálicas/patología , Células Cultivadas , Modelos Animales de Enfermedad , Gliosis/patología , Gliosis/fisiopatología , Proteínas Hedgehog/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal , Corteza Somatosensorial/lesiones , Corteza Somatosensorial/patología , Heridas Punzantes
8.
Dev Biol ; 413(1): 86-103, 2016 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-26187199

RESUMEN

During development of the CNS, stem and progenitor cell proliferation, cell fate designation, and patterning decisions are tightly regulated by interdependent networks of key transcriptional regulators. In a genetic approach we analyzed divergent functionality of the PAI and RED sub-domains of the Pax6 Paired domain (PD) during progenitor zone formation, motor and interneuron development, and peripheral connectivity at distinct levels within the neural tube: within the hindbrain, mutation of the PAI sub-domain severely affected patterning of the p3 and pMN domains and establishment of the corresponding motor neurons. Exit point designation of hypoglossal axons was disturbed in embryos harboring either mutations in the PD sub-domains or containing a functional Pax6 Null allele. At brachial spinal levels, we propose a selective involvement of the PAI sub-domain during patterning of ventral p2 and pMN domains, critically disturbing generation of specific motor neuron subtypes and increasing V2 interneuron numbers. Our findings present a novel aspect of how Pax6 not only utilizes its modular structure to perform distinct functions via its paired and homeodomain. Individual sub-domains can exert distinct functions, generating a new level of complexity for transcriptional regulation by one single transcription factor not only in dorso-ventral, but also rostro-caudal neural tube patterning.


Asunto(s)
Proteínas del Ojo/genética , Proteínas de Homeodominio/genética , Tubo Neural/embriología , Factores de Transcripción Paired Box/genética , Sistema Nervioso Periférico/embriología , Proteínas Represoras/genética , Alelos , Animales , Axones/metabolismo , Axones/fisiología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Tipificación del Cuerpo , Linaje de la Célula , Proliferación Celular , Proteínas de Unión al ADN/genética , Proteínas del Ojo/fisiología , Regulación del Desarrollo de la Expresión Génica , Genotipo , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/fisiología , Inmunohistoquímica , Hibridación in Situ , Interneuronas/metabolismo , Ratones , Neuronas Motoras/metabolismo , Mutación , Proteínas del Tejido Nervioso/genética , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/fisiología , Fenotipo , Estructura Terciaria de Proteína , Proteínas Represoras/fisiología , Rombencéfalo/metabolismo , Células Madre/citología , Factores de Transcripción/genética
9.
Glia ; 65(6): 990-1004, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28317180

RESUMEN

Astrocytes are the most abundant cell type of the central nervous system and cover a broad range of functionalities. We report here the generation of a novel monoclonal antibody, anti-astrocyte cell surface antigen-2 (Anti-ACSA-2). Flow cytometry, immunohistochemistry and immunocytochemistry revealed that Anti-ACSA-2 reacted specifically with a not yet identified glycosylated surface molecule of murine astrocytes at all developmental stages. It did not show any labeling of non-astroglial cells such as neurons, oligodendrocytes, NG2+ cells, microglia, endothelial cells, leukocytes, or erythrocytes. Co-labeling studies of GLAST and ACSA-2 showed largely overlapping expression. However, there were also notable differences in protein expression levels and frequencies of single-positive subpopulations of cells in some regions of the CNS such as cerebellum, most prominently at early postnatal stages. In the neurogenic niches, the dentate gyrus of the hippocampus and the subventricular zone (SVZ), again a general overlap with slight differences in expression levels were observed. ACSA-2 was unlike GLAST not sensitive to papain-based tissue dissociation and allowed for a highly effective, acute, specific, and prospective purification of viable astrocytes based on a new rapid sorting procedure using Anti-ACSA-2 directly coupled to superparamagnetic MicroBeads. In conclusion, ACSA-2 appears to be a new surface marker for astrocytes, radial glia, neural stem cells and bipotent glial progenitor cells which opens up the possibility of further dissecting the characteristics of astroglial subpopulations and lineages.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Antígenos de Superficie/análisis , Antígenos de Superficie/inmunología , Astrocitos/citología , Astrocitos/inmunología , Separación Inmunomagnética/métodos , Animales , Animales Recién Nacidos , Especificidad de Anticuerpos , Antígenos de Superficie/metabolismo , Encéfalo/citología , Encéfalo/crecimiento & desarrollo , Células Cultivadas , Células Endoteliales/citología , Células Endoteliales/inmunología , Eritrocitos/citología , Eritrocitos/metabolismo , Transportador 1 de Aminoácidos Excitadores/análisis , Leucocitos/citología , Leucocitos/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/citología , Microglía/inmunología , Células-Madre Neurales/inmunología , Neuronas/citología , Neuronas/metabolismo , Oligodendroglía/citología , Oligodendroglía/inmunología , Ratas Wistar
10.
Proteomics ; 16(3): 437-47, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26593131

RESUMEN

Histone posttranslational modifications and histone variants control the epigenetic regulation of gene expression and affect a wide variety of biological processes. A complex pattern of such modifications and variants defines the identity of cells within complex organ systems and can therefore be used to characterize cells at a molecular level. However, their detection and identification in situ has been limited so far due to lack of specificity, selectivity, and availability of antihistone antibodies. Here, we describe a novel MALDI imaging MS based workflow, which enables us to detect and characterize histones by their intact mass and their correlation with cytological properties of the tissue using novel statistical and image analysis tools. The workflow allows us to characterize the in situ distribution of the major histone variants and their modification in the mouse brain. This new analysis tool is particularly useful for the investigation of expression patterns of the linker histone H1 variants for which suitable antibodies are so far not available.


Asunto(s)
Encéfalo/metabolismo , Cromatina/química , Epigénesis Genética , Histonas/genética , Procesamiento Proteico-Postraduccional , Acetilación , Animales , Encéfalo/ultraestructura , Química Encefálica , Cromatina/metabolismo , Histonas/metabolismo , Masculino , Metilación , Ratones , Imagen Molecular/métodos , Fosforilación , Análisis de Componente Principal , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Biología de Sistemas/instrumentación , Biología de Sistemas/métodos
11.
Development ; 140(5): 1123-36, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23404109

RESUMEN

To achieve adequate organ development and size, cell proliferation and differentiation have to be tightly regulated and coordinated. The transcription factor Pax6 regulates patterning, neurogenesis and proliferation in forebrain development. The molecular basis of this regulation is not well understood. As the bipartite DNA-binding paired domain of Pax6 regulates forebrain development, we examined mice with point mutations in its individual DNA-binding subdomains PAI (Pax6(Leca4), N50K) and RED (Pax6(Leca2), R128C). This revealed distinct roles in regulating proliferation in the developing cerebral cortex, with the PAI and RED subdomain mutations reducing and increasing, respectively, the number of mitoses. Conversely, neurogenesis was affected only by the PAI subdomain mutation, phenocopying the neurogenic defects observed in full Pax6 mutants. Genome-wide expression profiling identified molecularly discrete signatures of Pax6(Leca4) and Pax6(Leca2) mutations. Comparison to Pax6 targets identified by chromatin immunoprecipitation led to the identification and functional characterization of distinct DNA motifs in the promoters of target genes dysregulated in the Pax6(Leca2) or Pax6(Leca4) mutants, further supporting the distinct regulatory functions of the DNA-binding subdomains. Thus, Pax6 achieves its key roles in the developing forebrain by utilizing particular subdomains to coordinate patterning, neurogenesis and proliferation simultaneously.


Asunto(s)
Proliferación Celular , Proteínas del Ojo/química , Proteínas del Ojo/genética , Proteínas del Ojo/fisiología , Proteínas de Homeodominio/química , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Neurogénesis/genética , Factores de Transcripción Paired Box/química , Factores de Transcripción Paired Box/genética , Factores de Transcripción Paired Box/fisiología , Proteínas Represoras/química , Proteínas Represoras/genética , Proteínas Represoras/fisiología , Animales , Animales Modificados Genéticamente , Tipificación del Cuerpo/genética , Tipificación del Cuerpo/fisiología , Embrión de Mamíferos , Proteínas del Ojo/metabolismo , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Modelos Biológicos , Mutagénesis Sitio-Dirigida , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/metabolismo , Prosencéfalo/embriología , Prosencéfalo/metabolismo , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína/genética , Estructura Terciaria de Proteína/fisiología , Proteínas Represoras/metabolismo , Transducción de Señal/genética
12.
Cell Tissue Res ; 359(1): 5-16, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25416507

RESUMEN

Neuronal differentiation relies on a set of interconnected molecular events to achieve the differentiation of pan-neuronal hallmarks, together with neuronal subtype-specific features. Here, we propose a conceptual framework for these events, based on recent findings. This framework encompasses a dimension in time during development, progressing from early master regulators to later expressed effector genes and terminal selector genes. As a horizontal intersection, we propose the action of permissive fate determinants that are critical in allowing progression through the above transcriptional phases. Typically, these are widely expressed and often interact with the chromatin remodeling machinery. We conclude by discussing this model in the context of the direct fate conversion of various somatic cells into neurons.


Asunto(s)
Sistema Nervioso Central/citología , Sistema Nervioso Central/metabolismo , Neurogénesis , Neuronas/citología , Animales , Linaje de la Célula , Humanos , Transcripción Genética
13.
Bioessays ; 35(3): 242-52, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23335359

RESUMEN

In the adult mammalian brain, neurogenesis is restricted to few regions, while gliogenesis continues in a wide-spread manner. Here we discuss our knowledge of extrinsic and intrinsic factors regulating neuro- and gliogenesis in the adult brain and propose a model of fate specification identifying the states of easiest transition between glio- and neurogenesis, highlighting the unique mechanisms stabilising the neural stem cell state. The model also encompasses the fate alterations achieved by direct reprogramming, and hence addresses a novel avenue for repair, namely eliciting neurogenesis from glial cells outside the neurogenic niches.


Asunto(s)
Encéfalo/citología , Linaje de la Célula , Neurogénesis , Neuroglía/citología , Adulto , Animales , Humanos , Modelos Biológicos , Nicho de Células Madre
14.
Development ; 138(23): 5067-78, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22031545

RESUMEN

Successful brain development requires tight regulation of sequential symmetric and asymmetric cell division. Although Pax6 is known to exert multiple roles in the developing nervous system, its role in the regulation of cell division is unknown. Here, we demonstrate profound alterations in the orientation and mode of cell division in the cerebral cortex of mice deficient in Pax6 function (Pax6(Sey/Sey)) or after acute induced deletion of Pax6. Live imaging revealed an increase in non-vertical cellular cleavage planes, resulting in an increased number of progenitors with unequal inheritance of the apical membrane domain and adherens junctions in the absence of Pax6 function. This phenotype appears to be mediated by the direct Pax6 target Spag5, a microtubule-associated protein, reduced levels of which result in the replication of the Pax6 phenotype of altered cell division orientation. In addition, lack of Pax6 also results in premature delamination of progenitor cells from the apical surface due to an overall decrease in proteins mediating anchoring at the ventricular surface. Moreover, continuous long-term imaging in vitro revealed that Pax6-deficient progenitors generate daughter cells with asymmetric fates at higher frequencies. These data demonstrate a cell-autonomous role for Pax6 in regulating the mode of cell division independently of apicobasal polarity and cell-cell interactions. Taken together, our work reveals several direct effects that the transcription factor Pax6 has on the machinery that mediates the orientation and mode of cell division.


Asunto(s)
División Celular/fisiología , Polaridad Celular/fisiología , Corteza Cerebral/citología , Corteza Cerebral/embriología , Proteínas del Ojo/metabolismo , Proteínas de Homeodominio/metabolismo , Factores de Transcripción Paired Box/metabolismo , Proteínas Represoras/metabolismo , Células Madre/fisiología , Análisis de Varianza , Animales , Proteínas de Ciclo Celular , Inmunoprecipitación de Cromatina , Cartilla de ADN/genética , Electroporación , Femenino , Inmunohistoquímica , Luciferasas , Ratones , Factor de Transcripción PAX6 , Reacción en Cadena de la Polimerasa/métodos , Embarazo
15.
Nat Commun ; 15(1): 2866, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38570482

RESUMEN

Traumatic brain injury leads to a highly orchestrated immune- and glial cell response partially responsible for long-lasting disability and the development of secondary neurodegenerative diseases. A holistic understanding of the mechanisms controlling the responses of specific cell types and their crosstalk is required to develop an efficient strategy for better regeneration. Here, we combine spatial and single-cell transcriptomics to chart the transcriptomic signature of the injured male murine cerebral cortex, and identify specific states of different glial cells contributing to this signature. Interestingly, distinct glial cells share a large fraction of injury-regulated genes, including inflammatory programs downstream of the innate immune-associated pathways Cxcr3 and Tlr1/2. Systemic manipulation of these pathways decreases the reactivity state of glial cells associated with poor regeneration. The functional relevance of the discovered shared signature of glial cells highlights the importance of our resource enabling comprehensive analysis of early events after brain injury.


Asunto(s)
Lesiones Encefálicas , Heridas Punzantes , Animales , Ratones , Masculino , Proteína Ácida Fibrilar de la Glía/metabolismo , Neuroglía/metabolismo , Lesiones Encefálicas/metabolismo , Corteza Cerebral/metabolismo , Heridas Punzantes/complicaciones , Heridas Punzantes/metabolismo
16.
Dev Cell ; 58(13): 1153-1169.e5, 2023 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-37220747

RESUMEN

Acute brain injuries evoke various response cascades directing the formation of the glial scar. Here, we report that acute lesions associated with hemorrhagic injuries trigger a re-programming of oligodendrocytes. Single-cell RNA sequencing highlighted a subpopulation of oligodendrocytes activating astroglial genes after acute brain injuries. By using PLP-DsRed1/GFAP-EGFP and PLP-EGFPmem/GFAP-mRFP1 transgenic mice, we visualized this population of oligodendrocytes that we termed AO cells based on their concomitant activity of astro- and oligodendroglial genes. By fate mapping using PLP- and GFAP-split Cre complementation and repeated chronic in vivo imaging with two-photon laser-scanning microscopy, we observed the conversion of oligodendrocytes into astrocytes via the AO cell stage. Such conversion was promoted by local injection of IL-6 and was diminished by IL-6 receptor-neutralizing antibody as well as by inhibiting microglial activation with minocycline. In summary, our findings highlight the plastic potential of oligodendrocytes in acute brain trauma due to microglia-derived IL-6.


Asunto(s)
Astrocitos , Lesiones Encefálicas , Ratones , Animales , Interleucina-6 , Proteína Ácida Fibrilar de la Glía/genética , Oligodendroglía , Ratones Transgénicos
17.
Nat Med ; 29(12): 3149-3161, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38066208

RESUMEN

The glial environment influences neurological disease progression, yet much of our knowledge still relies on preclinical animal studies, especially regarding astrocyte heterogeneity. In murine models of traumatic brain injury, beneficial functions of proliferating reactive astrocytes on disease outcome have been unraveled, but little is known regarding if and when they are present in human brain pathology. Here we examined a broad spectrum of pathologies with and without intracerebral hemorrhage and found a striking correlation between lesions involving blood-brain barrier rupture and astrocyte proliferation that was further corroborated in an assay probing for neural stem cell potential. Most importantly, proteomic analysis unraveled a crucial signaling pathway regulating this astrocyte plasticity with GALECTIN3 as a novel marker for proliferating astrocytes and the GALECTIN3-binding protein LGALS3BP as a functional hub mediating astrocyte proliferation and neurosphere formation. Taken together, this work identifies a therapeutically relevant astrocyte response and their molecular regulators in different pathologies affecting the human cerebral cortex.


Asunto(s)
Astrocitos , Células-Madre Neurales , Humanos , Ratones , Animales , Astrocitos/patología , Proteómica , Encéfalo , Sistema Nervioso Central
18.
Glia ; 60(3): 343-57, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22105794

RESUMEN

Reactive glia, including astroglia and oligodendrocyte progenitors (OPCs) are at the core of the reaction to injury in the mammalian brain with initially beneficial and later partially adverse functions such as scar formation. Given the different glial composition in the adult zebrafish brain with radial ependymoglia but no parenchymal astrocytes, we examined the glial response to an invasive stab wound injury model in the adult zebrafish telencephalon. Strikingly, already a few days after injury the wound was closed without any scar tissue. Similar to mammals, microglia cells reacted first and accumulated close to the injury site, while neither GFAP+ radial ependymoglia nor adult OPCs were recruited to the injury site. Moreover, OPCs failed to increase their proliferation after this injury, while the number of proliferating GFAP+ glia was increased until 7 days after injury. Importantly, neurogenesis was also increased after injury, generating additional neurons recruited to the parenchyma which survived for several months. Thus, these data suggest that the specific glial environment in the adult zebrafish telencephalon is not only permissive for long-term neuronal survival, but avoids scar formation. Invasive injury in the adult zebrafish telencephalon may therefore provide a useful model to untangle the molecular mechanisms involved in these beneficial glial reactions.


Asunto(s)
Modelos Animales de Enfermedad , Gliosis/etiología , Telencéfalo/lesiones , Heridas Punzantes/complicaciones , Aminoácidos , Animales , Animales Modificados Genéticamente , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Bromodesoxiuridina/metabolismo , Proliferación Celular , Regulación de la Expresión Génica/fisiología , Proteína Ácida Fibrilar de la Glía/genética , Proteínas Fluorescentes Verdes/genética , Microglía/clasificación , Microglía/patología , Proteínas del Tejido Nervioso/genética , Neurogénesis , Factor de Transcripción 2 de los Oligodendrocitos , Proteína Proto-Oncogénica c-fli-1/genética , Heridas Punzantes/patología , Pez Cebra , Proteínas de Pez Cebra/genética
19.
Curr Biol ; 32(23): R1314-R1316, 2022 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-36473442

RESUMEN

A new study finds the spliceosome protein SNRNP70 in cytoplasmic RNA granules in zebrafish motoneurons. Intriguingly, cytoplasmic SNRNP70 is essential for functional neuromuscular junctions, possibly due to a role in alternative splicing of z+agrin mRNA.


Asunto(s)
Empalme Alternativo , ARN , Animales , Pez Cebra/genética
20.
Cells ; 11(22)2022 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-36429113

RESUMEN

Peritubular cells of the human testis form a small compartment surrounding the seminiferous tubules. They are crucial for sperm transport, and they emerge as contributors to the spermatogonial stem cell niche. They are among the least known cell types of the human body. We employed single-cell RNA sequencing of cultured human testicular peritubular cells (HTPCs), which had been isolated from testicular samples of donors with normal spermatogenesis. The significant overlap between our results and recently published ex vivo data indicates that HTPCs are a highly adequate cellular model to define and study these cells. Thus, based on the expression of several markers, HTPCs can be classified as testicular smooth muscle cells. Small differences between the in vivo/in vitro expressed genes may be due to cellular plasticity. Plasticity was also shown upon addition of FCS to the culture medium. Based on transcriptome similarities, four cellular states were identified. Further analyses confirmed the presence of known stem cell niche-relevant factors (e.g., GDNF) and identified unknown functions, e.g., the ability to produce retinoic acid. Therefore, HTPCs allow us to define the signature(s) and delineate the functions of human testicular peritubular cells. The data may also serve as a resource for future studies to better understand male (in)fertility.


Asunto(s)
Análisis de la Célula Individual , Testículo , Humanos , Masculino , Testículo/metabolismo , Semen , Túbulos Seminíferos/metabolismo , Espermatogonias/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA