Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 130
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 174(4): 999-1014.e22, 2018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-30096314

RESUMEN

The mammalian nervous system executes complex behaviors controlled by specialized, precisely positioned, and interacting cell types. Here, we used RNA sequencing of half a million single cells to create a detailed census of cell types in the mouse nervous system. We mapped cell types spatially and derived a hierarchical, data-driven taxonomy. Neurons were the most diverse and were grouped by developmental anatomical units and by the expression of neurotransmitters and neuropeptides. Neuronal diversity was driven by genes encoding cell identity, synaptic connectivity, neurotransmission, and membrane conductance. We discovered seven distinct, regionally restricted astrocyte types that obeyed developmental boundaries and correlated with the spatial distribution of key glutamate and glycine neurotransmitters. In contrast, oligodendrocytes showed a loss of regional identity followed by a secondary diversification. The resource presented here lays a solid foundation for understanding the molecular architecture of the mammalian nervous system and enables genetic manipulation of specific cell types.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Sistema Nervioso/metabolismo , Análisis de la Célula Individual/métodos , Transcriptoma , Animales , Femenino , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Masculino , Ratones , Ratones Endogámicos C57BL , Sistema Nervioso/crecimiento & desarrollo
2.
Cell ; 167(2): 566-580.e19, 2016 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-27716510

RESUMEN

Understanding human embryonic ventral midbrain is of major interest for Parkinson's disease. However, the cell types, their gene expression dynamics, and their relationship to commonly used rodent models remain to be defined. We performed single-cell RNA sequencing to examine ventral midbrain development in human and mouse. We found 25 molecularly defined human cell types, including five subtypes of radial glia-like cells and four progenitors. In the mouse, two mature fetal dopaminergic neuron subtypes diversified into five adult classes during postnatal development. Cell types and gene expression were generally conserved across species, but with clear differences in cell proliferation, developmental timing, and dopaminergic neuron development. Additionally, we developed a method to quantitatively assess the fidelity of dopaminergic neurons derived from human pluripotent stem cells, at a single-cell level. Thus, our study provides insight into the molecular programs controlling human midbrain development and provides a foundation for the development of cell replacement therapies.


Asunto(s)
Neuronas Dopaminérgicas/citología , Mesencéfalo/citología , Mesencéfalo/embriología , Células-Madre Neurales/citología , Neurogénesis , Células Madre Pluripotentes/citología , Animales , Línea Celular , Técnicas de Reprogramación Celular , Humanos , Aprendizaje Automático , Mesencéfalo/metabolismo , Ratones , Neuroglía/citología , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos
3.
Cell ; 157(2): 313-328, 2014 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-24656405

RESUMEN

Glioblastoma multiforme (GBM) is the most aggressive form of brain cancer with marginal life expectancy. Based on the assumption that GBM cells gain functions not necessarily involved in the cancerous process, patient-derived glioblastoma cells (GCs) were screened to identify cellular processes amenable for development of targeted treatments. The quinine-derivative NSC13316 reliably and selectively compromised viability. Synthetic chemical expansion reveals delicate structure-activity relationship and analogs with increased potency, termed Vacquinols. Vacquinols stimulate death by membrane ruffling, cell rounding, massive macropinocytic vacuole accumulation, ATP depletion, and cytoplasmic membrane rupture of GCs. The MAP kinase MKK4, identified by a shRNA screen, represents a critical signaling node. Vacquinol-1 displays excellent in vivo pharmacokinetics and brain exposure, attenuates disease progression, and prolongs survival in a GBM animal model. These results identify a vulnerability to massive vacuolization that can be targeted by small molecules and point to the possible exploitation of this process in the design of anticancer therapies.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Piperidinas/farmacología , Quinolinas/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Muerte Celular/efectos de los fármacos , Xenoinjertos , Humanos , Hidroxiquinolinas/farmacología , MAP Quinasa Quinasa 4/metabolismo , Ratones , Trasplante de Neoplasias , Pinocitosis/efectos de los fármacos , Vacuolas/metabolismo , Pez Cebra
5.
Development ; 148(10)2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-34032267

RESUMEN

The choroid plexus (ChP) produces cerebrospinal fluid and forms an essential brain barrier. ChP tissues form in each brain ventricle, each one adopting a distinct shape, but remarkably little is known about the mechanisms underlying ChP development. Here, we show that epithelial WNT5A is crucial for determining fourth ventricle (4V) ChP morphogenesis and size in mouse. Systemic Wnt5a knockout, or forced Wnt5a overexpression beginning at embryonic day 10.5, profoundly reduced ChP size and development. However, Wnt5a expression was enriched in Foxj1-positive epithelial cells of 4V ChP plexus, and its conditional deletion in these cells affected the branched, villous morphology of the 4V ChP. We found that WNT5A was enriched in epithelial cells localized to the distal tips of 4V ChP villi, where WNT5A acted locally to activate non-canonical WNT signaling via ROR1 and ROR2 receptors. During 4V ChP development, MEIS1 bound to the proximal Wnt5a promoter, and gain- and loss-of-function approaches demonstrated that MEIS1 regulated Wnt5a expression. Collectively, our findings demonstrate a dual function of WNT5A in ChP development and identify MEIS transcription factors as upstream regulators of Wnt5a in the 4V ChP epithelium.


Asunto(s)
Plexo Coroideo/embriología , Epitelio/metabolismo , Cuarto Ventrículo/embriología , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/metabolismo , Proteína Wnt-5a/metabolismo , Animales , Encéfalo/embriología , Sistemas CRISPR-Cas/genética , Línea Celular , Células Epiteliales/metabolismo , Femenino , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas/genética , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , Transducción de Señal/fisiología , Proteína Wnt-5a/genética
6.
Int J Mol Sci ; 25(1)2024 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-38203854

RESUMEN

Mutations in the GBA1 gene increase the risk of developing Parkinson's disease (PD). However, most carriers of GBA1 mutations do not develop PD throughout their lives. The mechanisms of how GBA1 mutations contribute to PD pathogenesis remain unclear. Cerebrospinal fluid (CSF) is used for detecting pathological conditions of diseases, providing insights into the molecular mechanisms underlying neurodegenerative disorders. In this study, we utilized the proximity extension assay to examine the levels of metabolism-linked protein in the CSF from 17 PD patients carrying GBA1 mutations (GBA1-PD) and 17 idiopathic PD (iPD). The analysis of CSF secretome in GBA1-PD identified 11 significantly altered proteins, namely FKBP4, THOP1, GLRX, TXNDC5, GAL, SEMA3F, CRKL, APLP1, LRP11, CD164, and NPTXR. To investigate GBA1-associated CSF changes attributed to specific neuronal subtypes responsible for PD, we analyzed the cell culture supernatant from GBA1-PD-induced pluripotent stem cell (iPSC)-derived midbrain dopaminergic (mDA) neurons. The secretome analysis of GBA1-PD iPSC-derived mDA neurons revealed that five differently regulated proteins overlapped with those identified in the CSF analysis: FKBP4, THOP1, GLRX, GAL, and CRKL. Reduced intracellular level of the top hit, FKPB4, was confirmed via Western Blot. In conclusion, our findings identify significantly altered CSF GBA1-PD-associated proteins with FKPB4 being firmly attributed to mDA neurons.


Asunto(s)
Células Madre Pluripotentes Inducidas , Enfermedad de Parkinson , Proteínas de Unión a Tacrolimus , Humanos , Proteínas del Líquido Cefalorraquídeo , Proteínas de la Membrana , Mutación , Proteínas del Tejido Nervioso , Enfermedad de Parkinson/genética , Proteína Disulfuro Isomerasas , Secretoma , Proteínas de Unión a Tacrolimus/genética
7.
Development ; 146(16)2019 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-31371375

RESUMEN

Development of the central nervous system requires coordination of the proliferation and differentiation of neural stem cells. Here, we show that laminin alpha 2 (lm-α2) is a component of the midbrain dopaminergic neuron (mDA) progenitor niche in the ventral midbrain (VM) and identify a concentration-dependent role for laminin α2ß1γ1 (lm211) in regulating mDA progenitor proliferation and survival via a distinct set of receptors. At high concentrations, lm211-rich environments maintain mDA progenitors in a proliferative state via integrins α6ß1 and α7ß1, whereas low concentrations of lm211 support mDA lineage survival via dystroglycan receptors. We confirmed our findings in vivo, demonstrating that the VM was smaller in the absence of lm-α2, with increased apoptosis; furthermore, the progenitor pool was depleted through premature differentiation, resulting in fewer mDA neurons. Examination of mDA neuron subtype composition showed a reduction in later-born mDA neurons of the ventral tegmental area, which control a range of cognitive behaviours. Our results identify a novel role for laminin in neural development and provide a possible mechanism for autism-like behaviours and the brainstem hypoplasia seen in some individuals with mutations of LAMA2.


Asunto(s)
Neuronas Dopaminérgicas/fisiología , Laminina/fisiología , Mesencéfalo/embriología , Neurogénesis , Animales , Línea Celular , Proliferación Celular , Supervivencia Celular , Humanos , Integrinas/metabolismo , Laminina/genética , Mesencéfalo/citología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neurogénesis/genética
8.
9.
J Biol Chem ; 294(11): 4169-4176, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30655290

RESUMEN

The liver X receptors Lxrα/NR1H3 and Lxrß/NR1H2 are ligand-dependent nuclear receptors critical for midbrain dopaminergic (mDA) neuron development. We found previously that 24(S),25-epoxycholesterol (24,25-EC), the most potent and abundant Lxr ligand in the developing mouse midbrain, promotes mDA neurogenesis in vitro In this study, we demonstrate that 24,25-EC promotes mDA neurogenesis in an Lxr-dependent manner in the developing mouse midbrain in vivo and also prevents toxicity induced by the Lxr inhibitor geranylgeranyl pyrophosphate. Furthermore, using MS, we show that overexpression of human cholesterol 24S-hydroxylase (CYP46A1) increases the levels of both 24(S)-hydroxycholesterol (24-HC) and 24,25-EC in the developing midbrain, resulting in a specific increase in mDA neurogenesis in vitro and in vivo, but has no effect on oculomotor or red nucleus neurogenesis. 24-HC, unlike 24,25-EC, did not affect in vitro neurogenesis, indicating that the neurogenic effect of 24,25-EC on mDA neurons is specific. Combined, our results indicate that increased levels of 24,25-EC in vivo, by intracerebroventricular delivery in WT mice or by overexpression of its biosynthetic enzyme CYP46A1, specifically promote mDA neurogenesis. We propose that increasing the levels of 24,25-EC in vivo may be a useful strategy to combat the loss of mDA neurons in Parkinson's disease.


Asunto(s)
Colesterol 24-Hidroxilasa/biosíntesis , Colesterol/análogos & derivados , Dopamina/metabolismo , Mesencéfalo/metabolismo , Neurogénesis , Animales , Células Cultivadas , Colesterol/biosíntesis , Femenino , Humanos , Ratones , Ratones Transgénicos
10.
EMBO J ; 35(18): 1963-78, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27354364

RESUMEN

Pre-B-cell leukemia homeobox (PBX) transcription factors are known to regulate organogenesis, but their molecular targets and function in midbrain dopaminergic neurons (mDAn) as well as their role in neurodegenerative diseases are unknown. Here, we show that PBX1 controls a novel transcriptional network required for mDAn specification and survival, which is sufficient to generate mDAn from human stem cells. Mechanistically, PBX1 plays a dual role in transcription by directly repressing or activating genes, such as Onecut2 to inhibit lateral fates during embryogenesis, Pitx3 to promote mDAn development, and Nfe2l1 to protect from oxidative stress. Notably, PBX1 and NFE2L1 levels are severely reduced in dopaminergic neurons of the substantia nigra of Parkinson's disease (PD) patients and decreased NFE2L1 levels increases damage by oxidative stress in human midbrain cells. Thus, our results reveal novel roles for PBX1 and its transcriptional network in mDAn development and PD, opening the door for new therapeutic interventions.


Asunto(s)
Diferenciación Celular , Proteínas de Unión al ADN/metabolismo , Neuronas Dopaminérgicas/fisiología , Redes Reguladoras de Genes , Enfermedad de Parkinson/patología , Proteínas Proto-Oncogénicas/metabolismo , Sustancia Negra/patología , Humanos , Factor de Transcripción 1 de la Leucemia de Células Pre-B
12.
Development ; 142(11): 1918-36, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-26015536

RESUMEN

Midbrain dopaminergic (mDA) neuron development has been an intense area of research during recent years. This is due in part to a growing interest in regenerative medicine and the hope that treatment for diseases affecting mDA neurons, such as Parkinson's disease (PD), might be facilitated by a better understanding of how these neurons are specified, differentiated and maintained in vivo. This knowledge might help to instruct efforts to generate mDA neurons in vitro, which holds promise not only for cell replacement therapy, but also for disease modeling and drug discovery. In this Primer, we will focus on recent developments in understanding the molecular mechanisms that regulate the development of mDA neurons in vivo, and how they have been used to generate human mDA neurons in vitro from pluripotent stem cells or from somatic cells via direct reprogramming. Current challenges and future avenues in the development of a regenerative medicine for PD will be identified and discussed.


Asunto(s)
Neuronas Dopaminérgicas/citología , Mesencéfalo/citología , Neurogénesis , Animales , Tipificación del Cuerpo/genética , Neuronas Dopaminérgicas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Modelos Biológicos , Neurogénesis/genética
13.
Bioinformatics ; 33(16): 2547-2554, 2017 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-28430858

RESUMEN

MOTIVATION: Signal transduction via calcium ions (Ca2+) represents a fundamental signaling pathway in all eukaryotic cells. A large portion of the human genome encodes proteins used to assemble signaling systems that can transduce signals with diverse spatial and temporal dynamics. RESULTS: Here, we provide a map of all of the genes involved in Ca2+ signaling and link these genes to human genetic disorders. Using Gene Ontology terms and genome databases, 1805 genes were identified as regulators or targets of intracellular Ca2+ signals. Associating these 1805 genes with human genetic disorders uncovered 1470 diseases with mutated 'Ca2+ genes'. A network with scale-free properties appeared when the Ca2+ genes were mapped to their associated genetic disorders. AVAILABILITY AND IMPLEMENTATION: The Ca2+ genome database is freely available at http://cagedb.uhlenlab.org and will foster studies of gene functions and genetic disorders associated with Ca2+ signaling. CONTACT: per.uhlen@ki.se. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Señalización del Calcio/genética , Mapeo Cromosómico/métodos , Bases de Datos Genéticas , Genoma Humano , Genómica/métodos , Genética Humana/métodos , Humanos
14.
J Neurosci ; 35(39): 13385-401, 2015 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-26424886

RESUMEN

Wingless-related MMTV integration site 1 (WNT1)/ß-catenin signaling plays a crucial role in the generation of mesodiencephalic dopaminergic (mdDA) neurons, including the substantia nigra pars compacta (SNc) subpopulation that preferentially degenerates in Parkinson's disease (PD). However, the precise functions of WNT1/ß-catenin signaling in this context remain unknown. Stem cell-based regenerative (transplantation) therapies for PD have not been implemented widely in the clinical context, among other reasons because of the heterogeneity and incomplete differentiation of the transplanted cells. This might result in tumor formation and poor integration of the transplanted cells into the dopaminergic circuitry of the brain. Dickkopf 3 (DKK3) is a secreted glycoprotein implicated in the modulation of WNT/ß-catenin signaling. Using mutant mice, primary ventral midbrain cells, and pluripotent stem cells, we show that DKK3 is necessary and sufficient for the correct differentiation of a rostrolateral mdDA neuron subset. Dkk3 transcription in the murine ventral midbrain coincides with the onset of mdDA neurogenesis and is required for the activation and/or maintenance of LMX1A (LIM homeobox transcription factor 1α) and PITX3 (paired-like homeodomain transcription factor 3) expression in the corresponding mdDA precursor subset, without affecting the proliferation or specification of their progenitors. Notably, the treatment of differentiating pluripotent stem cells with recombinant DKK3 and WNT1 proteins also increases the proportion of mdDA neurons with molecular SNc DA cell characteristics in these cultures. The specific effects of DKK3 on the differentiation of rostrolateral mdDA neurons in the murine ventral midbrain, together with its known prosurvival and anti-tumorigenic properties, make it a good candidate for the improvement of regenerative and neuroprotective strategies in the treatment of PD. Significance statement: We show here that Dickkopf 3 (DKK3), a secreted modulator of WNT (Wingless-related MMTV integration site)/ß-catenin signaling, is both necessary and sufficient for the proper differentiation and survival of a rostrolateral (parabrachial pigmented nucleus and dorsomedial substantia nigra pars compacta) mesodiencephalic dopaminergic neuron subset, using Dkk3 mutant mice and murine primary ventral midbrain and pluripotent stem cells. The progressive loss of these dopamine-producing mesodiencephalic neurons is a hallmark of human Parkinson's disease, which can up to now not be halted by clinical treatments of this disease. Thus, the soluble DKK3 protein might be a promising new agent for the improvement of current protocols for the directed differentiation of pluripotent and multipotent stem cells into mesodiencephalic dopaminergic neurons and for the promotion of their survival in situ.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/fisiología , Mesencéfalo/fisiología , Células-Madre Neurales/fisiología , Células Madre Pluripotentes/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Recuento de Células , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Supervivencia Celular/genética , Células Cultivadas , Desoxiuridina/análogos & derivados , Desoxiuridina/farmacología , Péptidos y Proteínas de Señalización Intercelular/genética , Mesencéfalo/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transcriptoma , Proteína Wnt1/genética , Proteína Wnt1/fisiología
15.
Development ; 140(22): 4554-64, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24154522

RESUMEN

CXCL12/CXCR4 signaling has been reported to regulate three essential processes for the establishment of neural networks in different neuronal systems: neuronal migration, cell positioning and axon wiring. However, it is not known whether it regulates the development of A9-A10 tyrosine hydroxylase positive (TH(+)) midbrain dopaminergic (mDA) neurons. We report here that Cxcl12 is expressed in the meninges surrounding the ventral midbrain (VM), whereas CXCR4 is present in NURR1(+) mDA precursors and mDA neurons from E10.5 to E14.5. CXCR4 is activated in NURR1(+) cells as they migrate towards the meninges. Accordingly, VM meninges and CXCL12 promoted migration and neuritogenesis of TH(+) cells in VM explants in a CXCR4-dependent manner. Moreover, in vivo electroporation of Cxcl12 at E12.5 in the basal plate resulted in lateral migration, whereas expression in the midline resulted in retention of TH(+) cells in the IZ close to the midline. Analysis of Cxcr4(-/-) mice revealed the presence of VM TH(+) cells with disoriented processes in the intermediate zone (IZ) at E11.5 and marginal zone (MZ) at E14. Consistently, pharmacological blockade of CXCR4 or genetic deletion of Cxcr4 resulted in an accumulation of TH(+) cells in the lateral aspect of the IZ at E14, indicating that CXCR4 is required for the radial migration of mDA neurons in vivo. Altogether, our findings demonstrate that CXCL12/CXCR4 regulates the migration and orientation of processes in A9-A10 mDA neurons.


Asunto(s)
Movimiento Celular , Quimiocina CXCL12/metabolismo , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/metabolismo , Receptores CXCR4/metabolismo , Transducción de Señal , Animales , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Femenino , Eliminación de Gen , Masculino , Meninges/citología , Meninges/metabolismo , Mesencéfalo/citología , Mesencéfalo/embriología , Mesencéfalo/metabolismo , Ratones , Ratones Mutantes , Neuritas/metabolismo , Neurogénesis , Fosforilación , Tirosina 3-Monooxigenasa/metabolismo
16.
Proc Natl Acad Sci U S A ; 110(7): E602-10, 2013 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-23324743

RESUMEN

Wnts are a family of secreted proteins that regulate multiple steps of neural development and stem cell differentiation. Two of them, Wnt1 and Wnt5a, activate distinct branches of Wnt signaling and individually regulate different aspects of midbrain dopaminergic (DA) neuron development. However, several of their functions and interactions remain to be elucidated. Here, we report that loss of Wnt1 results in loss of Lmx1a and Ngn2 expression, as well as agenesis of DA neurons in the midbrain floor plate. Remarkably, a few ectopic DA neurons still emerge in the basal plate of Wnt1(-/-) mice, where Lmx1a is ectopically expressed. These results indicate that Wnt1 orchestrates DA specification and neurogenesis in vivo. Analysis of Wnt1(-/-);Wnt5a(-/-) mice revealed a greater loss of Nurr1(+) cells and DA neurons than in single mutants, indicating that Wnt1 and Wnt5a interact genetically and cooperate to promote midbrain DA neuron development in vivo. Our results unravel a functional interaction between Wnt1 and Wnt5a resulting in enhanced DA neurogenesis. Taking advantage of these findings, we have developed an application of Wnts to improve the generation of midbrain DA neurons from neural and embryonic stem cells. We thus show that coordinated Wnt actions promote DA neuron development in vivo and in stem cells and suggest that coordinated Wnt administration can be used to improve DA differentiation of stem cells and the development of stem cell-based therapies for Parkinson's disease.


Asunto(s)
Neuronas Dopaminérgicas/fisiología , Mesencéfalo/crecimiento & desarrollo , Neurogénesis/fisiología , Células Madre/citología , Proteínas Wnt/metabolismo , Vía de Señalización Wnt/fisiología , Proteína Wnt1/metabolismo , Análisis de Varianza , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Recuento de Células , Diferenciación Celular/fisiología , Neuronas Dopaminérgicas/metabolismo , Inmunohistoquímica , Proteínas con Homeodominio LIM/metabolismo , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/terapia , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Proteína Wnt-5a , Proteína Wnt1/deficiencia
17.
Proc Natl Acad Sci U S A ; 110(16): E1524-32, 2013 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-23576737

RESUMEN

Coherent network activity among assemblies of interconnected cells is essential for diverse functions in the adult brain. However, cellular networks before formations of chemical synapses are poorly understood. Here, embryonic stem cell-derived neural progenitors were found to form networks exhibiting synchronous calcium ion (Ca(2+)) activity that stimulated cell proliferation. Immature neural cells established circuits that propagated electrical signals between neighboring cells, thereby activating voltage-gated Ca(2+) channels that triggered Ca(2+) oscillations. These network circuits were dependent on gap junctions, because blocking prevented electrotonic transmission both in vitro and in vivo. Inhibiting connexin 43 gap junctions abolished network activity, suppressed proliferation, and affected embryonic cortical layer formation. Cross-correlation analysis revealed highly correlated Ca(2+) activities in small-world networks that followed a scale-free topology. Graph theory predicts that such network designs are effective for biological systems. Taken together, these results demonstrate that immature cells in the developing brain organize in small-world networks that critically regulate neural progenitor proliferation.


Asunto(s)
Encéfalo/embriología , Proliferación Celular , Red Nerviosa , Células-Madre Neurales/fisiología , Animales , Calcio/metabolismo , Canales de Calcio Tipo L/metabolismo , Conexina 43/metabolismo , Sinapsis Eléctricas/fisiología , Ratones , Ratones Endogámicos C57BL , Microscopía de Interferencia , Modelos Neurológicos , Células-Madre Neurales/citología , Plásmidos/genética , ARN Interferente Pequeño/genética
18.
Nat Rev Neurosci ; 11(2): 77-86, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20010950

RESUMEN

The roles of the Wnt signalling pathway in several developmental processes, including synaptic differentiation, are well characterized. The expression of Wnt ligands and Wnt signalling components in the mature mammalian CNS suggests that this pathway might also play a part in synaptic maintenance and function. In fact, Wnts have a crucial role in synaptic physiology, as they modulate the synaptic vesicle cycle, the trafficking of neurotransmitter receptors and the interaction of these receptors with scaffold proteins in postsynaptic regions. In addition, Wnts participate in adult neurogenesis and protect excitatory synaptic terminals from amyloid-beta oligomer toxicity. Here, the latest insights into the function of Wnt signalling in the adult nervous system and therapeutic opportunities for neurodegenerative diseases such as Alzheimer's and Parkinson's disease are discussed.


Asunto(s)
Fenómenos Fisiológicos del Sistema Nervioso , Proteínas Wnt/fisiología , Adulto , Factores de Edad , Animales , Sistema Nervioso Central/fisiología , Humanos , Transducción de Señal/fisiología
19.
Nat Chem Biol ; 9(2): 126-33, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23292650

RESUMEN

Liver X receptors (Lxrα and Lxrß) are ligand-dependent nuclear receptors critical for ventral midbrain neurogenesis in vivo. However, no endogenous midbrain Lxr ligand has so far been identified. Here we used LC/MS and functional assays to identify cholic acid as a new Lxr ligand. Moreover, 24(S),25-epoxycholesterol (24,25-EC) was found to be the most potent and abundant Lxr ligand in the developing mouse midbrain. Both Lxr ligands promoted neural development in an Lxr-dependent manner in zebrafish in vivo. Notably, each ligand selectively regulated the development of distinct midbrain neuronal populations. Whereas cholic acid increased survival and neurogenesis of Brn3a-positive red nucleus neurons, 24,25-EC promoted dopaminergic neurogenesis. These results identify an entirely new class of highly selective and cell type-specific regulators of neurogenesis and neuronal survival. Moreover, 24,25-EC promoted dopaminergic differentiation of embryonic stem cells, suggesting that Lxr ligands may thus contribute to the development of cell replacement and regenerative therapies for Parkinson's disease.


Asunto(s)
Mesencéfalo/metabolismo , Neurogénesis , Receptores Nucleares Huérfanos/metabolismo , Animales , Mapeo Encefálico/métodos , Diferenciación Celular , Núcleo Celular/metabolismo , Colesterol/análogos & derivados , Colesterol/metabolismo , Ácido Cólico/metabolismo , Dopamina/metabolismo , Relación Dosis-Respuesta a Droga , Células Madre Embrionarias/citología , Ligandos , Receptores X del Hígado , Ratones , Modelos Biológicos , Factores de Tiempo , Transfección , Pez Cebra
20.
Nature ; 451(7177): 460-4, 2008 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-18185516

RESUMEN

Stem cell self-renewal implies proliferation under continued maintenance of multipotency. Small changes in numbers of stem cells may lead to large differences in differentiated cell numbers, resulting in significant physiological consequences. Proliferation is typically regulated in the G1 phase, which is associated with differentiation and cell cycle arrest. However, embryonic stem (ES) cells may lack a G1 checkpoint. Regulation of proliferation in the 'DNA damage' S/G2 cell cycle checkpoint pathway is known for its role in the maintenance of chromatin structural integrity. Here we show that autocrine/paracrine gamma-aminobutyric acid (GABA) signalling by means of GABA(A) receptors negatively controls ES cell and peripheral neural crest stem (NCS) cell proliferation, preimplantation embryonic growth and proliferation in the boundary-cap stem cell niche, resulting in an attenuation of neuronal progenies from this stem cell niche. Activation of GABA(A) receptors leads to hyperpolarization, increased cell volume and accumulation of stem cells in S phase, thereby causing a rapid decrease in cell proliferation. GABA(A) receptors signal through S-phase checkpoint kinases of the phosphatidylinositol-3-OH kinase-related kinase family and the histone variant H2AX. This signalling pathway critically regulates proliferation independently of differentiation, apoptosis and overt damage to DNA. These results indicate the presence of a fundamentally different mechanism of proliferation control in these stem cells, in comparison with most somatic cells, involving proteins in the DNA damage checkpoint pathway.


Asunto(s)
Histonas/metabolismo , Receptores de GABA-A/metabolismo , Células Madre/citología , Células Madre/metabolismo , Animales , Comunicación Autocrina , Blastocisto/citología , Blastocisto/enzimología , Blastocisto/metabolismo , Recuento de Células , Ciclo Celular , Línea Celular , Proliferación Celular , Tamaño de la Célula , Daño del ADN , Agonistas de Receptores de GABA-A , Antagonistas de Receptores de GABA-A , Histonas/deficiencia , Histonas/genética , Ratones , Cresta Neural/citología , Cresta Neural/metabolismo , Comunicación Paracrina , Técnicas de Placa-Clamp , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Receptores de GABA-A/genética , Células Madre/enzimología , Ácido gamma-Aminobutírico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA