Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Proc Natl Acad Sci U S A ; 117(26): 15085-15095, 2020 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-32546527

RESUMEN

Comparative transcriptomics between differentiating human pluripotent stem cells (hPSCs) and developing mouse neurons offers a powerful approach to compare genetic and epigenetic pathways in human and mouse neurons. To analyze human Purkinje cell (PC) differentiation, we optimized a protocol to generate human pluripotent stem cell-derived Purkinje cells (hPSC-PCs) that formed synapses when cultured with mouse cerebellar glia and granule cells and fired large calcium currents, measured with the genetically encoded calcium indicator jRGECO1a. To directly compare global gene expression of hPSC-PCs with developing mouse PCs, we used translating ribosomal affinity purification (TRAP). As a first step, we used Tg(Pcp2-L10a-Egfp) TRAP mice to profile actively transcribed genes in developing postnatal mouse PCs and used metagene projection to identify the most salient patterns of PC gene expression over time. We then created a transgenic Pcp2-L10a-Egfp TRAP hPSC line to profile gene expression in differentiating hPSC-PCs, finding that the key gene expression pathways of differentiated hPSC-PCs most closely matched those of late juvenile mouse PCs (P21). Comparative bioinformatics identified classical PC gene signatures as well as novel mitochondrial and autophagy gene pathways during the differentiation of both mouse and human PCs. In addition, we identified genes expressed in hPSC-PCs but not mouse PCs and confirmed protein expression of a novel human PC gene, CD40LG, expressed in both hPSC-PCs and native human cerebellar tissue. This study therefore provides a direct comparison of hPSC-PC and mouse PC gene expression and a robust method for generating differentiated hPSC-PCs with human-specific gene expression for modeling developmental and degenerative cerebellar disorders.


Asunto(s)
Diferenciación Celular , Células de Purkinje/metabolismo , Transcriptoma , Animales , Humanos , Ratones , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Proteínas/genética , Proteínas/metabolismo , Células de Purkinje/citología
2.
Proc Natl Acad Sci U S A ; 115(42): 10556-10563, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-30262652

RESUMEN

Prior studies demonstrate that astrotactin (ASTN1) provides a neuronal receptor for glial-guided CNS migration. Here we report that ASTN1 binds N-cadherin (CDH2) and that the ASTN1:CDH2 interaction supports cell-cell adhesion. To test the function of ASTN1:CDH2 binding in glial-guided neuronal migration, we generated a conditional loss of Cdh2 in cerebellar granule cells and in glia. Granule cell migration was slowed in cerebellar slice cultures after a conditional loss of neuronal Cdh2, and more severe migration defects occurred after a conditional loss of glial Cdh2 Expression in granule cells of a mutant form of ASTN1 that does not bind CDH2 also slowed migration. Moreover, in vitro chimeras of granule cells and glia showed impaired neuron-glia attachment in the absence of glial, but not neuronal, Cdh2 Thus, cis and trans bindings of ASTN1 to neuronal and glial CDH2 form an asymmetric neuron-glial bridge complex that promotes glial-guided neuronal migration.


Asunto(s)
Cadherinas/fisiología , Adhesión Celular , Movimiento Celular , Cerebelo/fisiología , Glicoproteínas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuroglía/fisiología , Neuronas/fisiología , Animales , Células Cultivadas , Cerebelo/citología , Glicoproteínas/genética , Ligandos , Proteínas del Tejido Nervioso/genética , Neurogénesis , Neuroglía/citología , Neuronas/citología
3.
Proc Natl Acad Sci U S A ; 115(41): E9717-E9726, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30242134

RESUMEN

Surface protein dynamics dictate synaptic connectivity and function in neuronal circuits. ASTN2, a gene disrupted by copy number variations (CNVs) in neurodevelopmental disorders, including autism spectrum, was previously shown to regulate the surface expression of ASTN1 in glial-guided neuronal migration. Here, we demonstrate that ASTN2 binds to and regulates the surface expression of multiple synaptic proteins in postmigratory neurons by endocytosis, resulting in modulation of synaptic activity. In cerebellar Purkinje cells (PCs), by immunogold electron microscopy, ASTN2 localizes primarily to endocytic and autophagocytic vesicles in the cell soma and in subsets of dendritic spines. Overexpression of ASTN2 in PCs, but not of ASTN2 lacking the FNIII domain, recurrently disrupted by CNVs in patients, including in a family presented here, increases inhibitory and excitatory postsynaptic activity and reduces levels of ASTN2 binding partners. Our data suggest a fundamental role for ASTN2 in dynamic regulation of surface proteins by endocytic trafficking and protein degradation.


Asunto(s)
Variaciones en el Número de Copia de ADN , Glicoproteínas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Trastornos del Neurodesarrollo/genética , Sinapsis/fisiología , Animales , Movimiento Celular , Células Cultivadas , Endocitosis , Glicoproteínas/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Trastornos del Neurodesarrollo/patología , Transporte de Proteínas , Proteolisis , Células de Purkinje/metabolismo
4.
J Biol Chem ; 294(12): 4538-4545, 2019 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-30696770

RESUMEN

Astrotactin 1 (Astn1) and Astn2 are membrane proteins that function in glial-guided migration, receptor trafficking, and synaptic plasticity in the brain as well as in planar polarity pathways in the skin. Here we used glycosylation mapping and protease protection approaches to map the topologies of mouse Astn1 and Astn2 in rough microsomal membranes and found that Astn2 has a cleaved N-terminal signal peptide, an N-terminal domain located in the lumen of the rough microsomal membranes (topologically equivalent to the extracellular surface in cells), two transmembrane helices, and a large C-terminal lumenal domain. We also found that Astn1 has the same topology as Astn2, but we did not observe any evidence of signal peptide cleavage in Astn1. Both Astn1 and Astn2 mature through endoproteolytic cleavage in the second transmembrane helix; importantly, we identified the endoprotease responsible for the maturation of Astn1 and Astn2 as the endoplasmic reticulum signal peptidase. Differences in the degree of Astn1 and Astn2 maturation possibly contribute to the higher levels of the C-terminal domain of Astn1 detected on neuronal membranes of the central nervous system. These differences may also explain the distinct cellular functions of Astn1 and Astn2, such as in membrane adhesion, receptor trafficking, and planar polarity signaling.


Asunto(s)
Glicoproteínas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Biocatálisis , Retículo Endoplásmico/metabolismo , Glicoproteínas/química , Glicosilación , Membranas Intracelulares/metabolismo , Ratones , Microsomas/metabolismo , Proteínas del Tejido Nervioso/química , Proteolisis
5.
Elife ; 102021 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-34842137

RESUMEN

Brain development is regulated by conserved transcriptional programs across species, but little is known about the divergent mechanisms that create species-specific characteristics. Among brain regions, human cerebellar histogenesis differs in complexity compared with nonhuman primates and rodents, making it important to develop methods to generate human cerebellar neurons that closely resemble those in the developing human cerebellum. We report a rapid protocol for the derivation of the human ATOH1 lineage, the precursor of excitatory cerebellar neurons, from human pluripotent stem cells (hPSCs). Upon transplantation into juvenile mice, hPSC-derived cerebellar granule cells migrated along glial fibers and integrated into the cerebellar cortex. By Translational Ribosome Affinity Purification-seq, we identified an unexpected temporal shift in the expression of RBFOX3 (NeuN) and NEUROD1, which are classically associated with differentiated neurons, in the human outer external granule layer. This molecular divergence may enable the protracted development of the human cerebellum compared to mice.


Asunto(s)
Antígenos Nucleares/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Cerebelo/metabolismo , Proteínas del Tejido Nervioso/genética , Animales , Antígenos Nucleares/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Línea Celular , Humanos , Ratones , Ratones Endogámicos C57BL , Mutación , Proteínas del Tejido Nervioso/metabolismo
6.
Dev Biol ; 333(2): 360-72, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19576202

RESUMEN

Tbx2 is a T-box transcription factor gene that is dynamically expressed in the presumptive retina during optic vesicle invagination. Several findings implicate Tbx2 in cell cycle regulation, including its overexpression in tumours and regulation of proliferation during heart development. We investigated the role of Tbx2 in optic cup formation by analysing mice with a targeted homozygous mutation in Tbx2. Loss of Tbx2 caused a reduced presumptive retinal volume due to increased apoptosis, and a delay in ventral optic vesicle invagination leading to the formation of small and abnormally shaped optic cups. Tbx2 is essential for maintenance, but not induction of expression of the dorsal retinal determinant, Tbx5, and acts downstream of Bmp4, a dorsally expressed gene implicated in human microphthalmia. The small retina showed a hypocellular ventral region, loss of Fgf15, normally expressed in proliferating central retinal cells, and increased numbers of mitotic cells in the dorsal region, indicating that Tbx2 is required for normal growth and development across the D-V axis. Dorsal expression of potential regulators of retinal growth, Cyp1b1 and Cx43, and the topographic guidance molecule ephrinB2, was increased, and intraretinal axons were disorganised resulting in a failure of optic nerve formation. Our data provide evidence that Tbx2 is required for proper optic cup formation and plays a critical early role in regulating regional retinal growth and the acquisition of shape during optic vesicle invagination.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Retina/embriología , Retina/metabolismo , Proteínas de Dominio T Box/fisiología , Animales , Apoptosis , Muerte Celular , Diferenciación Celular , Proliferación Celular , Ojo/embriología , Ojo/metabolismo , Femenino , Hibridación in Situ , Ratones , Modelos Biológicos , Mutación , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo
7.
Hum Mol Genet ; 17(22): 3446-58, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18694899

RESUMEN

The factors that mediate chromosomal rearrangement remain incompletely defined. Among regions prone to structural variant formation, chromosome 6p25 is one of the few in which disease-associated segmental duplications and segmental deletions have been identified, primarily through gene dosage attributable ocular phenotypes. Using array comparative genome hybridization, we studied ten 6p25 duplication and deletion pedigrees and amplified junction fragments from each. Analysis of the breakpoint architecture revealed that all the rearrangements were non-recurrent, and in contrast to most previous examples the majority of the segmental duplications and deletions utilized coupled homologous and non-homologous recombination mechanisms. One junction fragment exhibited an unprecedented 367 bp insert derived from tandemly arranged breakpoint elements. While this accorded with a recently described replication-based mechanism, it differed from the previous example in being unassociated with template switching, and occurring in a segmental deletion. These results extend the mechanisms involved in structural variant formation, provide strong evidence that a spectrum of recombination, DNA repair and replication underlie 6p25 rearrangements, and have implications for genesis of copy number variations in other genomic regions. These findings highlight the benefits of undertaking the extensive studies necessary to characterize structural variants at the base pair level.


Asunto(s)
Aberraciones Cromosómicas , Cromosomas Humanos Par 6/genética , Dosificación de Gen , Glaucoma/genética , Rotura Cromosómica , Deleción Cromosómica , Factores de Transcripción Forkhead , Haplotipos , Humanos , Hibridación de Ácido Nucleico , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Polimorfismo de Nucleótido Simple , Recombinación Genética
8.
BMC Dev Biol ; 6: 62, 2006 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17173667

RESUMEN

BACKGROUND: Polarised gene expression is thought to lead to the graded distribution of signaling molecules providing a patterning mechanism across the embryonic eye. Bone morphogenetic protein 4 (Bmp4) is expressed in the dorsal optic vesicle as it transforms into the optic cup. Bmp4 deletions in human and mouse result in failure of eye development, but little attempt has been made to investigate mammalian targets of BMP4 signaling. In chick, retroviral gene overexpression studies indicate that Bmp4 activates the dorsally expressed Tbx5 gene, which represses ventrally expressed cVax. It is not known whether the Tbx5 related genes, Tbx2 and Tbx3, are BMP4 targets in the mammalian retina and whether BMP4 acts at a distance from its site of expression. Although it is established that Drosophila Dpp (homologue of vertebrate Bmp4) acts as a morphogen, there is little evidence that BMP4 gradients are interpreted to create domains of BMP4 target gene expression in the mouse. RESULTS: Our data show that the level of BMP4 signaling is critical for the regulation of distinct Tbx2, Tbx3, Tbx5 and Vax2 gene expression domains along the dorso-ventral axis of the mouse optic cup. BMP4 signaling gradients were manipulated in whole mouse embryo cultures during optic cup development, by implantation of beads soaked in BMP4, or the BMP antagonist Noggin, to provide a local signaling source. Tbx2, Tbx3 and Tbx5, showed a differential response to alterations in the level of BMP4 along the entire dorso-ventral axis of the optic cup, suggesting that BMP4 acts across a distance. Increased levels of BMP4 caused expansion of Tbx2 and Tbx3, but not Tbx5, into the ventral retina and repression of the ventral marker Vax2. Conversely, Noggin abolished Tbx5 expression but only shifted Tbx2 expression dorsally. Increased levels of BMP4 signaling caused decreased proliferation, reduced retinal volume and altered the shape of the optic cup. CONCLUSION: Our findings suggest the existence of a dorsal-high, ventral-low BMP4 signaling gradient across which distinct domains of Tbx2, Tbx3, Tbx5 and Vax2 transcription factor gene expression are set up. Furthermore we show that the correct level of BMP4 signaling is critical for normal growth of the mammalian embryonic eye.


Asunto(s)
Tipificación del Cuerpo/genética , Proteínas Morfogenéticas Óseas/genética , Ojo/embriología , Regulación del Desarrollo de la Expresión Génica , Transducción de Señal , Proteínas de Dominio T Box/genética , Animales , Proteína Morfogenética Ósea 4 , Proliferación Celular , Ojo/citología , Femenino , Hibridación in Situ , Etiquetado Corte-Fin in Situ , Ratones , Ratones Endogámicos CBA , Embarazo
9.
Dis Model Mech ; 6(1): 49-63, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23065639

RESUMEN

BMI1 is a potent inducer of neural stem cell self-renewal and neural progenitor cell proliferation during development and in adult tissue homeostasis. It is overexpressed in numerous human cancers - including medulloblastomas, in which its functional role is unclear. We generated transgenic mouse lines with targeted overexpression of Bmi1 in the cerebellar granule cell lineage, a cell type that has been shown to act as a cell of origin for medulloblastomas. Overexpression of Bmi1 in granule cell progenitors (GCPs) led to a decrease in cerebellar size due to decreased GCP proliferation and repression of the expression of cyclin genes, whereas Bmi1 overexpression in postmitotic granule cells improved cell survival in response to stress by altering the expression of genes in the mitochondrial cell death pathway and of Myc and Lef-1. Although no medulloblastomas developed in ageing cohorts of transgenic mice, crosses with Trp53(-/-) mice resulted in a low incidence of medulloblastoma formation. Furthermore, analysis of a large collection of primary human medulloblastomas revealed that tumours with a BMI1(high) TP53(low) molecular profile are significantly enriched in Group 4 human medulloblastomas. Our data suggest that different levels and timing of Bmi1 overexpression yield distinct cellular outcomes within the same cellular lineage. Importantly, Bmi1 overexpression at the GCP stage does not induce tumour formation, suggesting that BMI1 overexpression in GCP-derived human medulloblastomas probably occurs during later stages of oncogenesis and might serve to enhance tumour cell survival.


Asunto(s)
Cerebelo/citología , Cerebelo/metabolismo , Complejo Represivo Polycomb 1/genética , Proteínas Proto-Oncogénicas/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Ciclo Celular , Linaje de la Célula , Proliferación Celular , Supervivencia Celular , Neoplasias Cerebelosas/etiología , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/patología , Epigénesis Genética , Femenino , Genes p53 , Humanos , Masculino , Meduloblastoma/etiología , Meduloblastoma/genética , Meduloblastoma/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Modelos Neurológicos , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Receptores Patched , Receptores de Superficie Celular/genética , Transcriptoma , Regulación hacia Arriba
10.
Int J Biochem Cell Biol ; 41(3): 435-45, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18755286

RESUMEN

Cerebellar granule cells originate from precursors located in the dorsal region of rhombomere one within the hindbrain of developing embryos. They undergo proliferation for an extensive period well into postnatal stages of development to form the major cell type of the cerebellum, the most populous structure within the mammalian brain. Granule cell development is highly dependent upon the cerebellar environment and contact with neighbouring cells. In recent years, the molecular basis of these interactions has started to be unravelled. Granule cell precursors and the molecular mechanisms involved in controlling their proliferation have been shown to be involved in the pathogenesis of medulloblastoma, the most common malignant pediatric brain tumour. Here, we review the control of granule cell generation with emphasis on the molecular regulators of cell proliferation and differentiation during normal and malignant development.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Neoplasias Cerebelosas/patología , Cerebelo/patología , Meduloblastoma/patología , Animales , Transformación Celular Neoplásica , Neoplasias Cerebelosas/etiología , Neoplasias Cerebelosas/metabolismo , Neoplasias Cerebelosas/fisiopatología , Cerebelo/embriología , Gránulos Citoplasmáticos/ultraestructura , Regulación del Desarrollo de la Expresión Génica , Humanos , Meduloblastoma/etiología , Meduloblastoma/metabolismo , Meduloblastoma/fisiopatología , Ratones , Neuronas/patología , Ratas , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA