Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Mol Neurosci ; 15: 988993, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36353360

RESUMEN

Mowat-Wilson syndrome (MWS) is a severe neurodevelopmental disorder caused by heterozygous variants in the gene encoding transcription factor ZEB2. Affected individuals present with structural brain abnormalities, speech delay and epilepsy. In mice, conditional loss of Zeb2 causes hippocampal degeneration, altered migration and differentiation of GABAergic interneurons, a heterogeneous population of mainly inhibitory neurons of importance for maintaining normal excitability. To get insights into GABAergic development and function in MWS we investigated ZEB2 haploinsufficient induced pluripotent stem cells (iPSC) of MWS subjects together with iPSC of healthy donors. Analysis of RNA-sequencing data at two time points of GABAergic development revealed an attenuated interneuronal identity in MWS subject derived iPSC with enrichment of differentially expressed genes required for transcriptional regulation, cell fate transition and forebrain patterning. The ZEB2 haploinsufficient neural stem cells (NSCs) showed downregulation of genes required for ventral telencephalon specification, such as FOXG1, accompanied by an impaired migratory capacity. Further differentiation into GABAergic interneuronal cells uncovered upregulation of transcription factors promoting pallial and excitatory neurons whereas cortical markers were downregulated. The differentially expressed genes formed a neural protein-protein network with extensive connections to well-established epilepsy genes. Analysis of electrophysiological properties in ZEB2 haploinsufficient GABAergic cells revealed overt perturbations manifested as impaired firing of repeated action potentials. Our iPSC model of ZEB2 haploinsufficient GABAergic development thus uncovers a dysregulated gene network leading to immature interneurons with mixed identity and altered electrophysiological properties, suggesting mechanisms contributing to the neuropathogenesis and seizures in MWS.

3.
Am J Hum Genet ; 108(4): 739-748, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33711248

RESUMEN

Neurochondrin (NCDN) is a cytoplasmatic neural protein of importance for neural growth, glutamate receptor (mGluR) signaling, and synaptic plasticity. Conditional loss of Ncdn in mice neural tissue causes depressive-like behaviors, impaired spatial learning, and epileptic seizures. We report on NCDN missense variants in six affected individuals with variable degrees of developmental delay, intellectual disability (ID), and seizures. Three siblings were found homozygous for a NCDN missense variant, whereas another three unrelated individuals carried different de novo missense variants in NCDN. We assayed the missense variants for their capability to rescue impaired neurite formation in human neuroblastoma (SH-SY5Y) cells depleted of NCDN. Overexpression of wild-type NCDN rescued the neurite-phenotype in contrast to expression of NCDN containing the variants of affected individuals. Two missense variants, associated with severe neurodevelopmental features and epilepsy, were unable to restore mGluR5-induced ERK phosphorylation. Electrophysiological analysis of SH-SY5Y cells depleted of NCDN exhibited altered membrane potential and impaired action potentials at repolarization, suggesting NCDN to be required for normal biophysical properties. Using available transcriptome data from human fetal cortex, we show that NCDN is highly expressed in maturing excitatory neurons. In combination, our data provide evidence that bi-allelic and de novo variants in NCDN cause a clinically variable form of neurodevelopmental delay and epilepsy, highlighting a critical role for NCDN in human brain development.


Asunto(s)
Alelos , Epilepsia/genética , Discapacidad Intelectual/genética , Mutación/genética , Proteínas del Tejido Nervioso/genética , Trastornos del Neurodesarrollo/genética , Adolescente , Secuencia de Bases , Línea Celular , Preescolar , Consanguinidad , Femenino , Humanos , Lactante , Trastornos del Desarrollo del Lenguaje/genética , Masculino , Mutación Missense , Neuritas , Pakistán
4.
Sci Rep ; 10(1): 20675, 2020 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-33244084

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a devastating incurable neurological disorder characterized by motor neuron (MN) death and muscle dysfunction leading to mean survival time after diagnosis of only 2-5 years. A potential ALS treatment is to delay the loss of MNs and disease progression by the delivery of trophic factors. Previously, we demonstrated that implanted mesoporous silica nanoparticles (MSPs) loaded with trophic factor peptide mimetics support survival and induce differentiation of co-implanted embryonic stem cell (ESC)-derived MNs. Here, we investigate whether MSP loaded with peptide mimetics of ciliary neurotrophic factor (Cintrofin), glial-derived neurotrophic factor (Gliafin), and vascular endothelial growth factor (Vefin1) injected into the cervical spinal cord of mutant SOD1 mice affect disease progression and extend survival. We also transplanted boundary cap neural crest stem cells (bNCSCs) which have been shown previously to have a positive effect on MN survival in vitro and in vivo. We show that mimetic-loaded MSPs and bNCSCs significantly delay disease progression and increase survival of mutant SOD1 mice, and also that empty particles significantly improve the condition of ALS mice. Our results suggest that intraspinal delivery of MSPs is a potential therapeutic approach for the treatment of ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Esclerosis Amiotrófica Lateral/patología , Supervivencia Celular/efectos de los fármacos , Dióxido de Silicio/farmacología , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Células Cultivadas , Médula Cervical/efectos de los fármacos , Médula Cervical/metabolismo , Médula Cervical/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/patología , Femenino , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Ratones , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Cresta Neural/efectos de los fármacos , Cresta Neural/metabolismo , Cresta Neural/patología , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
5.
Stem Cell Res ; 49: 102081, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33220594

RESUMEN

Down syndrome (DS) is caused by trisomy for chromosome 21 (T21). We generated two induced pluripotent stem cell (iPSC) lines from skin fibroblasts of two males with DS using Sendai virus delivery of OCT4, SOX2, KLF4, and c-MYC. Characterization of the two iPSC lines, UUIGPi013-A and UUIPGi014-A, showed that they are genetically stable with a 47,XY,+21 karyotype. Both lines displayed expression of pluripotency markers and trilineage differentiation capacity. These two iPSC lines provide a useful resource for DS modeling and pharmacological interventions.


Asunto(s)
Síndrome de Down , Células Madre Pluripotentes Inducidas , Diferenciación Celular , Cromosomas Humanos Par 21 , Síndrome de Down/genética , Humanos , Factor 4 Similar a Kruppel , Masculino , Trisomía/genética
6.
Int J Hematol ; 112(6): 894-899, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32772263

RESUMEN

Diamond-Blackfan Anemia (DBA) is a congenital pure red cell aplasia caused by heterozygous variants in ribosomal protein genes. The hematological features associated with DBA are highly variable and non-hematological abnormalities are common. We report herein on an affected mother and her daughter presenting with transfusion-dependent anemia. The mother showed mild physical abnormalities and entered spontaneous remission at age 13 years. Her daughter was born with occipital meningocele. Exome sequencing of DNA from the mother revealed a heterozygous novel splice site variant (NM_001011.4:c.508-3T > G) in the Ribosomal Protein S7 gene (RPS7) inherited by the daughter. Functional analysis of the RPS7 variant expressed from a mini-gene construct revealed that the exon 7 acceptor splice site was replaced by a cryptic splice resulting in a transcript missing 64 bp of exon 7 (p.Val170Serfs*8). Our study confirms a pathogenic effect of a novel RPS7 variant in DBA associated with spontaneous remission in the mother and meningocele in her daughter, thus adding to the genotype-phenotype correlations in DBA.


Asunto(s)
Anemia de Diamond-Blackfan/genética , Aberraciones Cromosómicas , Estudios de Asociación Genética , Variación Genética/genética , Meningocele/genética , Empalme del ARN/genética , Proteínas Ribosómicas/genética , Adolescente , Adulto , Anemia de Diamond-Blackfan/etiología , Niño , Exones/genética , Femenino , Humanos , Meningocele/etiología , Relaciones Madre-Hijo , Remisión Espontánea , Análisis de Secuencia de ADN
7.
ACS Chem Neurosci ; 11(9): 1270-1282, 2020 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-32283014

RESUMEN

Vascular endothelial growth factor B (VEGFB) is a pleiotropic trophic factor, which in contrast to the closely related VEGFA is known to have a limited effect on angiogenesis. VEGFB improves survival in various tissues including the nervous system, where the effect was observed mainly for peripheral neurons. The neurotrophic effect of VEGFB on central nervous system neurons has been less investigated. Here we demonstrated that VEGFB promotes neurite outgrowth from primary cerebellar granule, hippocampal, and retinal neurons in vitro. VEGFB protected hippocampal and retinal neurons from both oxidative stress and glutamate-induced neuronal death. The VEGF receptor 1 (VEGFR1) is required for VEGFB-induced neurotrophic and neuroprotective effects. Using a structure-based approach, we designed short peptides, termed Vefin1-7, mimicking the binding interface of VEGFB to VEGFR1. Vefins were analyzed for their secondary structure and binding to VEGF receptors and compared with previously described peptides derived from VEGFA, another ligand of VEGFR1. We show that Vefins have neurotrophic and neuroprotective effects on primary hippocampal, cerebellar granule, and retinal neurons in vitro with potencies comparable to VEGFB. Similar to VEGFB, Vefins were not mitogenic for MCF-7 cancer cells. Furthermore, one of the peptides, Vefin7, even dose-dependently inhibited the proliferation of MCF-7 cells in vitro. Unraveling the neurotrophic and neuroprotective potentials of VEGFB, the only nonangiogenic factor of the VEGF family, is promising for the development of neuroprotective peptide-based therapies.


Asunto(s)
Factor B de Crecimiento Endotelial Vascular , Receptor 1 de Factores de Crecimiento Endotelial Vascular , Sistema Nervioso Central , Neuronas , Péptidos/farmacología
8.
Stem Cell Res ; 44: 101739, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32126327

RESUMEN

Incontinentia pigmenti (IP) is an X-linked dominant neuroectodermal dysplasia caused by loss-of-function mutations in the IKBKG gene. Using CRISPR/Cas9 technology, we generated an IKBKG knock-out iPSC line (KICRi002-A-1) on a 46,XY background. The iPSC line showed a normal karyotype, expressed pluripotency markers and exhibited capability to differentiate into the three germ layers in vitro. Off-target editing was excluded and no IKBKG mRNA expression could be detected. Our line offers a useful resource to elucidate mechanisms caused by IKBKG deficiency that leads to disrupted male fetal development and for drug screening to improve treatment of female patients with IP.


Asunto(s)
Línea Celular , Incontinencia Pigmentaria , Células Madre Pluripotentes Inducidas , Sistemas CRISPR-Cas/genética , Femenino , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Incontinencia Pigmentaria/genética , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Mutación
9.
Stem Cell Res ; 44: 101758, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32203915

RESUMEN

The role of Neurochondrin (NCDN) in humans is not well understood. Mice with a conditional Ncdn knock-out show epileptic seizures, depressive-like behaviours and impaired spatial learning. Using CRISPR/Cas9, we generated a Neurochondrin deficient human iPSC line KICRi002-A-3 carrying a homozygous 752 bp deletion / 2 bp insertion in the NCDN gene. The iPSC line maintained a normal 46,XY karyotype, expressed pluripotency markers and exhibited capability to differentiate into the three germ layers in vitro. Off-target editing was excluded and Neurochondrin expression was not detectable. The iPSC line offers a valuable resource to study the role of Neurochondrin during human neurogenesis.


Asunto(s)
Células Madre Pluripotentes Inducidas , Animales , Sistemas CRISPR-Cas/genética , Línea Celular , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Humanos , Ratones , Proteínas del Tejido Nervioso
10.
Clin Epigenetics ; 12(1): 9, 2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31915063

RESUMEN

BACKGROUND: Down syndrome (DS) is characterized by neurodevelopmental abnormalities caused by partial or complete trisomy of human chromosome 21 (T21). Analysis of Down syndrome brain specimens has shown global epigenetic and transcriptional changes but their interplay during early neurogenesis remains largely unknown. We differentiated induced pluripotent stem cells (iPSCs) established from two DS patients with complete T21 and matched euploid donors into two distinct neural stages corresponding to early- and mid-gestational ages. RESULTS: Using the Illumina Infinium 450K array, we assessed the DNA methylation pattern of known CpG regions and promoters across the genome in trisomic neural iPSC derivatives, and we identified a total of 500 stably and differentially methylated CpGs that were annotated to CpG islands of 151 genes. The genes were enriched within the DNA binding category, uncovering 37 factors of importance for transcriptional regulation and chromatin structure. In particular, we observed regional epigenetic changes of the transcription factor genes ZNF69, ZNF700 and ZNF763 as well as the HOXA3, HOXB3 and HOXD3 genes. A similar clustering of differential methylation was found in the CpG islands of the HIST1 genes suggesting effects on chromatin remodeling. CONCLUSIONS: The study shows that early established differential methylation in neural iPSC derivatives with T21 are associated with a set of genes relevant for DS brain development, providing a novel framework for further studies on epigenetic changes and transcriptional dysregulation during T21 neurogenesis.


Asunto(s)
Encéfalo/metabolismo , Metilación de ADN/genética , Síndrome de Down/genética , Epigenómica/métodos , Células Madre Pluripotentes Inducidas/metabolismo , Adulto , Encéfalo/patología , Ensamble y Desensamble de Cromatina/genética , Islas de CpG/genética , Síndrome de Down/complicaciones , Femenino , Feto/metabolismo , Feto/patología , Regulación de la Expresión Génica/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Masculino , Trastornos del Neurodesarrollo/etiología , Trastornos del Neurodesarrollo/genética , Neurogénesis/genética , Embarazo , Regiones Promotoras Genéticas , Factores de Transcripción/genética , Trisomía/genética
11.
Front Cell Neurosci ; 13: 346, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31474832

RESUMEN

Mutations in superoxide dismutase (SOD1) are the second most common cause of familial amyotrophic lateral sclerosis (ALS), a fatal neurodegenerative disease caused by the death of motor neurons in the brain and spinal cord. SOD1 neurotoxicity has been attributed to aberrant accumulation of misfolded SOD1, which in its soluble form binds to intracellular organelles, such as mitochondria and ER, disrupting their functions. Here, we demonstrate that mutant SOD1 binds specifically to the N-terminal domain of the voltage-dependent anion channel (VDAC1), an outer mitochondrial membrane protein controlling cell energy, metabolic and survival pathways. Mutant SOD1G93A and SOD1G85R, but not wild type SOD1, directly interact with VDAC1 and reduce its channel conductance. No such interaction with N-terminal-truncated VDAC1 occurs. Moreover, a VDAC1-derived N-terminal peptide inhibited mutant SOD1-induced toxicity. Incubation of motor neuron-like NSC-34 cells expressing mutant SOD1 or mouse embryonic stem cell-derived motor neurons with different VDAC1 N-terminal peptides resulted in enhanced cell survival. Taken together, our results establish a direct link between mutant SOD1 toxicity and the VDAC1 N-terminal domain and suggest that VDAC1 N-terminal peptides targeting mutant SOD1 provide potential new therapeutic strategies for ALS.

12.
Mol Neurobiol ; 56(10): 7113-7127, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30989628

RESUMEN

Down syndrome (DS) or trisomy 21 (T21) is a leading genetic cause of intellectual disability. To gain insights into dynamics of molecular perturbations during neurogenesis in DS, we established a model using induced pluripotent stem cells (iPSC) with transcriptome profiles comparable to that of normal fetal brain development. When applied on iPSCs with T21, transcriptome and proteome signatures at two stages of differentiation revealed strong temporal dynamics of dysregulated genes, proteins and pathways belonging to 11 major functional clusters. DNA replication, synaptic maturation and neuroactive clusters were disturbed at the early differentiation time point accompanied by a skewed transition from the neural progenitor cell stage and reduced cellular growth. With differentiation, growth factor and extracellular matrix, oxidative phosphorylation and glycolysis emerged as major perturbed clusters. Furthermore, we identified a marked dysregulation of a set of genes encoded by chromosome 21 including an early upregulation of the hub gene APP, supporting its role for disturbed neurogenesis, and the transcription factors OLIG1, OLIG2 and RUNX1, consistent with deficient myelination and neuronal differentiation. Taken together, our findings highlight novel sequential and differentiation-dependent dynamics of disturbed functions, pathways and elements in T21 neurogenesis, providing further insights into developmental abnormalities of the DS brain.


Asunto(s)
Síndrome de Down/genética , Síndrome de Down/patología , Células Madre Pluripotentes Inducidas/patología , Neuronas/metabolismo , Neuronas/patología , Proteoma/metabolismo , Transcriptoma/genética , Diferenciación Celular/genética , Proliferación Celular/genética , Femenino , Humanos , Masculino , Mitocondrias/genética , Modelos Biológicos , Neuritas/metabolismo , Neurogénesis/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo , Transcripción Genética
13.
Regen Med ; 12(4): 339-351, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28621171

RESUMEN

AIM: During development, boundary cap neural crest stem cells (bNCSCs) assist sensory axon growth into the spinal cord. Here we repositioned them to test if they assist regeneration of sensory axons in adult mice after dorsal root avulsion injury. MATERIALS & METHODS: Avulsed mice received bNCSC or human neural progenitor (hNP) cell transplants and their contributions to glial scar formation and sensory axon regeneration were analyzed with immunohistochemistry and transganglionic tracing. RESULTS: hNPs and bNCSCs form similar gaps in the glial scar, but unlike hNPs, bNCSCs contribute Mts1/S100A4 (calcium-binding protein) expression to the scar and do not assist sensory axon regeneration. CONCLUSION: bNCSC transplants contribute nonpermissive Mts1/S100A4-expressing cells to the glial scar after dorsal root avulsion.


Asunto(s)
Cicatriz/patología , Cicatriz/terapia , Cresta Neural/trasplante , Trasplante de Células Madre , Animales , Astrocitos/metabolismo , Axones/patología , Biomarcadores/metabolismo , Línea Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Regeneración Nerviosa , Cresta Neural/citología , Proteína de Unión al Calcio S100A4/metabolismo , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/fisiopatología , Traumatismos de la Médula Espinal/terapia , Raíces Nerviosas Espinales/lesiones , Raíces Nerviosas Espinales/patología
14.
Stem Cells Dev ; 26(14): 1065-1077, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28562227

RESUMEN

Spinal root injuries result in newly formed glial scar formation, which prevents regeneration of sensory axons causing permanent sensory loss. Previous studies showed that delivery of trophic factors or implantation of human neural progenitor cells supports sensory axon regeneration and partly restores sensory functions. In this study, we elucidate mechanisms underlying stem cell-mediated ingrowth of sensory axons after dorsal root avulsion (DRA). We show that human spinal cord neural stem/progenitor cells (hscNSPC), and also, mesoporous silica particles loaded with growth factor mimetics (MesoMIM), supported sensory axon regeneration. However, when hscNSPC and MesoMIM were combined, sensory axon regeneration failed. Morphological and tracing analysis showed that sensory axons grow through the newly established glial scar along "bridges" formed by migrating stem cells. Coimplantation of MesoMIM prevented stem cell migration, "bridges" were not formed, and sensory axons failed to enter the spinal cord. MesoMIM applied alone supported sensory axons ingrowth, but without affecting glial scar formation. In vitro, the presence of MesoMIM significantly impaired migration of hscNSPC without affecting their level of differentiation. Our data show that (1) the ability of stem cells to migrate into the spinal cord and organize cellular "bridges" in the newly formed interface is crucial for successful sensory axon regeneration, (2) trophic factor mimetics delivered by mesoporous silica may be a convenient alternative way to induce sensory axon regeneration, and (3) a combinatorial approach of individually beneficial components is not necessarily additive, but can be counterproductive for axonal growth.


Asunto(s)
Axones/patología , Regeneración Nerviosa , Traumatismos de la Médula Espinal/fisiopatología , Médula Espinal/patología , Médula Espinal/fisiopatología , Raíces Nerviosas Espinales/patología , Raíces Nerviosas Espinales/fisiopatología , Animales , Diferenciación Celular , Movimiento Celular , Ganglión/patología , Humanos , Ratones , Células-Madre Neurales/trasplante , Neuroglía/patología , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/terapia , Trasplante de Células Madre
15.
Neurotherapeutics ; 14(3): 773-783, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28070746

RESUMEN

ALS is a devastating disease resulting in degeneration of motor neurons (MNs) in the brain and spinal cord. The survival of MNs strongly depends on surrounding glial cells and neurotrophic support from muscles. We previously demonstrated that boundary cap neural crest stem cells (bNCSCs) can give rise to neurons and glial cells in vitro and in vivo and have multiple beneficial effects on co-cultured and co-implanted cells, including neural cells. In this paper, we investigate if bNCSCs may improve survival of MNs harboring a mutant form of human SOD1 (SOD1G93A) in vitro under normal conditions and oxidative stress and in vivo after implantation to the spinal cord. We found that survival of SOD1G93A MNs in vitro was increased in the presence of bNCSCs under normal conditions as well as under oxidative stress. In addition, when SOD1G93A MN precursors were implanted to the spinal cord of adult mice, their survival was increased when they were co-implanted with bNCSCs. These findings show that bNCSCs support survival of SOD1G93A MNs in normal conditions and under oxidative stress in vitro and improve their survival in vivo, suggesting that bNCSCs have a potential for the development of novel stem cell-based therapeutic approaches in ALS models.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Neuronas Motoras/patología , Cresta Neural , Células-Madre Neurales , Animales , Supervivencia Celular , Células Cultivadas , Humanos , Ratones , Ratones Desnudos , Mutación , Cresta Neural/trasplante , Células-Madre Neurales/citología , Células-Madre Neurales/trasplante , Trasplante de Células Madre , Superóxido Dismutasa-1/genética
16.
Sci Rep ; 5: 10666, 2015 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-26053681

RESUMEN

Dorsal root avulsion results in permanent impairment of sensory functions due to disconnection between the peripheral and central nervous system. Improved strategies are therefore needed to reconnect injured sensory neurons with their spinal cord targets in order to achieve functional repair after brachial and lumbosacral plexus avulsion injuries. Here, we show that sensory functions can be restored in the adult mouse if avulsed sensory fibers are bridged with the spinal cord by human neural progenitor (hNP) transplants. Responses to peripheral mechanical sensory stimulation were significantly improved in transplanted animals. Transganglionic tracing showed host sensory axons only in the spinal cord dorsal horn of treated animals. Immunohistochemical analysis confirmed that sensory fibers had grown through the bridge and showed robust survival and differentiation of the transplants. Section of the repaired dorsal roots distal to the transplant completely abolished the behavioral improvement. This demonstrates that hNP transplants promote recovery of sensorimotor functions after dorsal root avulsion, and that these effects are mediated by spinal ingrowth of host sensory axons. These results provide a rationale for the development of novel stem cell-based strategies for functionally useful bridging of the peripheral and central nervous system.


Asunto(s)
Axones/fisiología , Células Madre Embrionarias Humanas/fisiología , Regeneración Nerviosa/fisiología , Células Receptoras Sensoriales/fisiología , Traumatismos de la Médula Espinal/fisiopatología , Raíces Nerviosas Espinales/fisiología , Células Madre/fisiología , Animales , Ganglios Espinales/fisiología , Humanos , Masculino , Ratones , Médula Espinal/fisiología
17.
Neural Regen Res ; 10(11): 1739-40, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26807098
18.
PLoS One ; 8(5): e63474, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23723986

RESUMEN

With remarkably few exceptions, the molecules mediating synaptic vesicle exocytosis at active zones are structurally and functionally conserved between vertebrates and invertebrates. Mover was found in a yeast-2-hybrid assay using the vertebrate-specific active zone scaffolding protein bassoon as a bait. Peptides of Mover have been reported in proteomics screens for self-interacting proteins, phosphorylated proteins, and synaptic vesicle proteins, respectively. Here, we tested the predictions arising from these screens. Using flotation assays, carbonate stripping of peripheral membrane proteins, mass spectrometry, immunogold labelling of purified synaptic vesicles, and immuno-organelle isolation, we found that Mover is indeed a peripheral synaptic vesicle membrane protein. In addition, by generating an antibody against phosphorylated Mover and Western blot analysis of fractionated rat brain, we found that Mover is a bona fide phospho-protein. The localization of Mover to synaptic vesicles is phosphorylation dependent; treatment with a phosphatase caused Mover to dissociate from synaptic vesicles. A yeast-2-hybrid screen, co-immunoprecipitation and cell-based optical assays of homomerization revealed that Mover undergoes homophilic interaction, and regions within both the N- and C- terminus of the protein are required for this interaction. Deleting a region required for homomeric interaction abolished presynaptic targeting of recombinant Mover in cultured neurons. Together, these data prove that Mover is associated with synaptic vesicles, and implicate phosphorylation and multimerization in targeting of Mover to synaptic vesicles and presynaptic sites.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Fosfoproteínas/metabolismo , Vesículas Sinápticas/metabolismo , Animales , Chlorocebus aethiops , Potenciales de la Membrana , Proteínas de la Membrana/metabolismo , Ratones , Proteínas Mutantes/metabolismo , Fosforilación , Unión Proteica , Transporte de Proteínas , Ratas , Ratas Sprague-Dawley , Eliminación de Secuencia , Fracciones Subcelulares/metabolismo , Vesículas Sinápticas/ultraestructura , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...