Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 124
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Exp Cell Res ; 438(1): 114049, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38642790

RESUMEN

BACKGROUND: Acellular nerve allografts (ANAs) have been successfully applied to bridge facial nerve defects, and transplantation of stem cells may enhance the regenerative results. Up to now, application of hair follicle epidermal neural crest stem cell-derived Schwann cell-like cells (EPI-NCSC-SCLCs) combined with ANAs for bridging facial nerve defects has not been reported. METHODS: The effect of ANAs laden with green fluorescent protein (GFP)-labeled EPI-NCSC-SCLCs (ANA + cells) on bridging rat facial nerve trunk defects (5-mm-long) was detected by functional and morphological examination, as compared with autografts and ANAs, respectively. RESULTS: (1) EPI-NCSC-SCLCs had good compatibility with ANAs in vitro. (2) In the ANA + cells group, the GFP signals were observed by in vivo imaging system for small animals within 8 weeks, and GFP-labeled EPI-NCSC-SCLCs were detected in the tissue slices at 16 weeks postoperatively. (3) The facial symmetry at rest after surgery in the ANA + cells group was better than that in the ANA group (p < 0.05), and similar to that in the autograft group (p > 0.05). The initial recovery time of vibrissal and eyelid movement in the ANA group was 2 weeks later than that in the other two groups. (4) The myelinated fibers, myelin sheath thickness and diameter of the axons of the buccal branches in the ANA group were significantly worse than those in the other two groups (P < 0.05), and the results in the ANA + cells group were similar to those in the autograft group (p > 0.05). CONCLUSIONS: EPI-NCSC-SCLCs could promote functional and morphological recovery of rat facial nerve defects, and GFP labeling could track the transplanted EPI-NCSC-SCLCs in vivo for a certain period of time. These may provide a novel choice for clinical treatment of peripheral nerve defects.


Asunto(s)
Aloinjertos , Nervio Facial , Proteínas Fluorescentes Verdes , Folículo Piloso , Regeneración Nerviosa , Cresta Neural , Células de Schwann , Animales , Células de Schwann/trasplante , Folículo Piloso/trasplante , Folículo Piloso/citología , Cresta Neural/citología , Cresta Neural/trasplante , Ratas , Proteínas Fluorescentes Verdes/metabolismo , Proteínas Fluorescentes Verdes/genética , Regeneración Nerviosa/fisiología , Células-Madre Neurales/trasplante , Células-Madre Neurales/citología , Ratas Sprague-Dawley , Traumatismos del Nervio Facial/terapia , Traumatismos del Nervio Facial/patología , Traumatismos del Nervio Facial/cirugía , Masculino
2.
Neurogastroenterol Motil ; 32(9): e13895, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32515097

RESUMEN

BACKGROUND: Hirschsprung's disease (HSCR) is a congenital gastrointestinal disorder, characterized by enteric ganglia absence in part or entire of the colon, due to abnormal colonization and migration of enteric neural crest cells (ENCCs) during development. Currently, besides surgery which is the main therapy for HSCR, the potential of stem cell-based transplantation was investigated as an alternative option. Although promising, it has limitations, including poor survival, differentiation, and migration of the grafted cells. We hypothesized that modulation of extracellular factors during transplantation could promote ENCCs proliferation and migration, leading to increased transplantation efficiency. Considering that the RhoA/ROCK pathway is highly involved in cytoskeletal dynamics and neurite growth, our study explored the effect of inhibition of this pathway to improve the success of ENCCs transplantation. METHODS: Enteric neural crest cells were isolated from rat embryos and labeled with a GFP-tag. Cell viability, apoptosis, differentiation, and migration assays were performed with and without RhoA/ROCK inhibition. Labeled ENCCs were transplanted into the muscle layer of an induced hypoganglionic rat model followed by intraperitoneal injections of ROCK inhibitor. The transplanted segments were collected 3 weeks after for histological analysis. KEY RESULTS: Our results showed that inhibition of ROCK increased viable cell number, differentiation, and migration of ENCCs in vitro. Moreover, transplantation of labeled ENCCs into the hypoganglionic model showed enhanced distribution of grafted ENCCs, upon treatment with ROCK inhibitor. CONCLUSIONS AND INFERENCES: ROCK inhibitors influence ENCCs growth and migration in vitro and in vivo, and should be considered to improve the efficiency of ENCCs transplantation.


Asunto(s)
Sistema Nervioso Entérico/metabolismo , Enfermedad de Hirschsprung/metabolismo , Cresta Neural/trasplante , Transducción de Señal/fisiología , Quinasas Asociadas a rho/metabolismo , Animales , Apoptosis/fisiología , Diferenciación Celular/fisiología , Supervivencia Celular/fisiología , Modelos Animales de Enfermedad , Enfermedad de Hirschsprung/patología , Ratas , Ratas Sprague-Dawley
3.
CNS Neurosci Ther ; 26(7): 670-681, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32281225

RESUMEN

INTRODUCTION: Cell-based therapy is considered as promising strategy to cure stroke. However, employing appropriate type of stem cell to fulfill many therapeutic needs of cerebral ischemia is still challenging. In this regard, the current study was designed to elucidate therapeutic potential of epidermal neural crest stem cells (EPI-NCSCs) compared to bone marrow mesenchymal stem cells (BM-MSCs) in rat model of ischemic stroke. METHODS: Ischemic stroke was induced by middle cerebral artery occlusion (MCAO) for 45 minutes. Immediately after reperfusion, EPI-NCSCs or BM-MSCs were transplanted via intra-arterial or intravenous route. A test for neurological function was performed before ischemia and 1, 3, and 7 days after MCAO. Also, infarct volume ratio and relative expression of 15 selected target genes were evaluated 7 days after transplantation. RESULTS: EPI-NCSCs transplantation (both intra-arterial and intravenous) and BM-MSCs transplantation (only intra-arterial) tended to result in a better functional outcome, compared to the MCAO group; however, this difference was not statistically significant. The infarct volume ratio significantly decreased in NCSC-intra-arterial, NCSC-intravenous and MSC-intra-arterial groups compared to the control. EPI-NCSCs interventions led to higher expression levels of Bdnf, nestin, Sox10, doublecortin, ß-III tubulin, Gfap, and interleukin-6, whereas neurotrophin-3 and interleukin-10 were decreased. On the other hand, BM-MSCs therapy resulted in upregulation of Gdnf, ß-III tubulin, and Gfap and down-regulation of neurotrophin-3, interleukin-1, and interleukin-10. CONCLUSION: These findings highlight the therapeutic effects of EPI-NCSCs transplantation, probably through simultaneous induction of neuronal and glial formation, as well as Bdnf over-expression in a rat model of ischemic stroke.


Asunto(s)
Isquemia Encefálica/terapia , Células Epidérmicas/trasplante , Accidente Cerebrovascular Isquémico/terapia , Cresta Neural/trasplante , Células-Madre Neurales/trasplante , Trasplante de Células Madre/métodos , Animales , Isquemia Encefálica/metabolismo , Proteína Doblecortina , Células Epidérmicas/metabolismo , Accidente Cerebrovascular Isquémico/metabolismo , Masculino , Cresta Neural/metabolismo , Células-Madre Neurales/metabolismo , Ratas , Ratas Sprague-Dawley
4.
Stem Cell Res Ther ; 10(1): 397, 2019 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-31852510

RESUMEN

BACKGROUND: Emerging evidence suggests that neural crest-derived cells (NCCs) present important functions in peripheral nerve regeneration to correct the insufficiency of autogenous Schwann cells. Postmigratory NCCs have been successfully isolated from adult rat bone marrow in our previous work. In this study, we aim to provide neural crest-derived Schwann cell precursors (SCPs) for repair of nerve defects in adult rats, and partially reveal the mechanisms involved in neuroregeneration of cell therapy. METHODS: A clonal cell line of neural crest precursors of rat bone marrow origin (rBM-NCPs) with SCP identity was expanded in adherent monolayer culture to ensure the stable cell viability of NCPs and potentiate the repair of nerve defects after rBM-NCPs implantation based on tissue engineering nerve grafts (TENG). Here the behavioral, morphological, and electrophysiological detection was performed to evaluate the therapy efficacy. We further investigated the treatment with NCP-conditioned medium (NCP-CM) to sensory neurons after exposure to oxygen-glucose-deprivation (OGD) and partially compared the expression of trophic factor genes in rBM-NCPs with that in mesenchymal stem cells of bone marrow origin (rBM-MSCs). RESULTS: It was showed that the constructed TENG with rBM-NCPs loaded into silk fibroin fiber scaffolds/chitosan conduits repaired 10-mm long sciatic nerve defects more efficiently than conduits alone. The axonal regrowth, remyelination promoted the reinnervation of the denervated hind limb muscle and skin and thereby alleviated muscle atrophy and facilitated the rehabilitation of motor and sensory function. Moreover, it was demonstrated that treatment with NCP-CM could restore the cultured primary sensory neurons after OGD through trophic factors including epidermal growth factor (EGF), platelet-derived growth factor alpha (PDGFα), ciliary neurotrophic factor (CNTF), and vascular endothelial growth factor alpha (VEGFα). CONCLUSIONS: In summary, our findings indicated that monolayer-cultured rBM-NCPs cell-based therapy might effectively repair peripheral nerve defects partially through secreted trophic factors, which represented the secretome of rBM-NCPs differing from that of rBM-MSCs.


Asunto(s)
Factor de Crecimiento Nervioso/metabolismo , Cresta Neural/trasplante , Traumatismos de los Nervios Periféricos/terapia , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Células de la Médula Ósea/citología , Diferenciación Celular , Quitosano/química , Fibroínas/química , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Regeneración Nerviosa , Cresta Neural/citología , Cresta Neural/metabolismo , Neuronas/metabolismo , Neuronas/patología , Traumatismos de los Nervios Periféricos/patología , Ratas , Ratas Wistar , Células de Schwann/citología , Células de Schwann/metabolismo , Ingeniería de Tejidos , Andamios del Tejido/química , Trasplante Autólogo
5.
Nat Commun ; 9(1): 4286, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-30327457

RESUMEN

A tissue engineered oesophagus could overcome limitations associated with oesophageal substitution. Combining decellularized scaffolds with patient-derived cells shows promise for regeneration of tissue defects. In this proof-of-principle study, a two-stage approach for generation of a bio-artificial oesophageal graft addresses some major challenges in organ engineering, namely: (i) development of multi-strata tubular structures, (ii) appropriate re-population/maturation of constructs before transplantation, (iii) cryopreservation of bio-engineered organs and (iv) in vivo pre-vascularization. The graft comprises decellularized rat oesophagus homogeneously re-populated with mesoangioblasts and fibroblasts for the muscle layer. The oesophageal muscle reaches organised maturation after dynamic culture in a bioreactor and functional integration with neural crest stem cells. Grafts are pre-vascularised in vivo in the omentum prior to mucosa reconstitution with expanded epithelial progenitors. Overall, our optimised two-stage approach produces a fully re-populated, structurally organized and pre-vascularized oesophageal substitute, which could become an alternative to current oesophageal substitutes.


Asunto(s)
Esófago/citología , Esófago/fisiología , Músculo Esquelético/citología , Ingeniería de Tejidos/métodos , Andamios del Tejido , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Niño , Preescolar , Criopreservación/métodos , Células Epiteliales , Matriz Extracelular/fisiología , Humanos , Lactante , Recién Nacido , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Cresta Neural/trasplante , Ratas Sprague-Dawley
6.
Integr Comp Biol ; 58(3): 411-420, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29718239

RESUMEN

Evolutionary changes in Astyanax mexicanus cavefish with respect to conspecific surface fish, including the regression of eyes, loss of pigmentation, and modification of the cranial skeleton, involve derivatives of the neural crest. However, the role of neural crest cells in cavefish evolution and development is poorly understood. One of the reasons is that experimental methods for neural crest analysis are not well developed in the Astyanax system. Here we describe neural crest transplantation between Astyanax surface fish and cavefish embryos. We found differences in the migration of cranial neural crest cells transplanted from the surface fish anterior hindbrain to the same region of surface fish or cavefish hosts. Cranial neural crest cells migrated extensively throughout the head, and to a lesser extent the trunk, in surface fish hosts but their migration was mostly restricted to the anterior and dorsal head regions in cavefish hosts. Cranial neural crest cells derived from the surface fish transplants invaded the degenerating eyes of cavefish hosts, resulting in increased eye size and suggesting that cavefish neural crest cells are defective in forming optic derivatives. We found that melanophores were formed in albino cavefish from grafts of surface fish trunk neural crest cells, showing that the cavefish tissue environment is conducive for pigment cell development, and implicating intrinsic changes in cavefish neural crest cells in loss of body pigmentation. It is concluded that changes in neural crest cells play key roles in the evolution of cavefish development.


Asunto(s)
Evolución Biológica , Characidae/embriología , Ojo/embriología , Cresta Neural/trasplante , Animales , Characidae/fisiología , Oscuridad , Luz , Fenómenos Fisiológicos Oculares
7.
Stem Cell Reports ; 10(5): 1445-1452, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29606614

RESUMEN

Chimeric mice have been generated by injecting pluripotent stem cells into morula-to-blastocyst stage mouse embryo or by introducing more mature cells into later stage embryos that correspond to the differentiation stage of the donor cells. It has not been rigorously tested, however, whether successful chimera formation requires the developmental stage of host embryo and donor cell to be matched. Here, we compared the success of chimera formation following injection of primary neural crest cells (NCCs) into blastocysts or of embryonic stem cells (ESCs) into E8.5 embryos (heterochronic injection) with that of injecting ESCs cells into the blastocyst or NCCs into the E8.5 embryos (isochronic injection). Chimera formation was efficient when donor and host were matched, but no functional chimeric contribution was found in heterochronic injections. This suggests that matching the developmental stage of donor cells with the host embryo is crucial for functional engraftment of donor cells into the developing embryo.


Asunto(s)
Quimera/metabolismo , Cresta Neural/citología , Animales , Apoptosis , Blastocisto/citología , Células Cultivadas , Embrión de Mamíferos/citología , Células Madre Embrionarias/citología , Células Madre Embrionarias/trasplante , Gastrulación , Inyecciones , Ratones Endogámicos C57BL , Cresta Neural/trasplante , Trasplante de Células Madre , Factores de Tiempo
8.
Eur J Pediatr Surg ; 28(3): 215-221, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29689583

RESUMEN

Stem cells possess the ability of self-renewal and the potency to differentiate into multiple cell lineages. Somatic stem cells are present in adult tissues, but they usually exhibit limited differentiation capacity and life span. On the other hand, somatic cells from adult tissues can be reprogrammed into induced pluripotent stem cells (iPSCs) that retain a full differentiation capacity with unlimited self-renewal ability. Autologous origin of iPSCs makes them an ideal source of cells for regenerative medicine to replenish the missing or damaged cells in the patients. iPSCs nowadays have also been widely used to build human disease models to study pathological mechanisms of the diseases. Hirschsprung disease (HSCR) is a congenital disorder caused by defects in the development of enteric neural crest stem cells. The failures of the ENCCs to proliferate, differentiate, and/or migrate lead to the absence of enteric neurons in the distal colon, resulting in colonic motility dysfunction. The lack of effective treatment for HSCR urges continuous efforts to develop new therapies for this congenital disorder. In this review, we will discuss the potential applications of somatic stem cells and iPSCs for the cell-based therapy of HSCR. We will also highlight the recent advances in stem cell research for the establishment of human HSCR models for the development of novel therapies.


Asunto(s)
Células Madre Adultas/trasplante , Enfermedad de Hirschsprung/terapia , Trasplante de Células Madre/métodos , Células Madre Embrionarias/trasplante , Humanos , Células Madre Pluripotentes Inducidas/trasplante , Cresta Neural/trasplante , Células-Madre Neurales/trasplante , Resultado del Tratamiento
9.
J Cell Mol Med ; 22(6): 3119-3132, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29536619

RESUMEN

Corneal opacities are a leading cause of global blindness. They are conventionally treated by the transplantation of donor corneal tissue, which is, restricted by a worldwide donor material shortage and allograft rejection. Autologous adult stem cells with a potential to differentiate into corneal stromal keratocytes (CSKs) could offer a suitable choice of cells for regenerative cell therapy. Postnatal periodontal ligament (PDL) contains a population of adult stem cells, which has a similar embryological origin as CSK, that is cranial neural crest. We harvested PDL cells from young adult teeth extracted because of non-functional or orthodontic reason and differentiated them towards CSK phenotype using a two-step protocol with spheroid formation followed by growth factor and cytokine induction in a stromal environment (human amnion stroma and porcine corneal stroma). Our results showed that the PDL-differentiated CSK-like cells expressed CSK markers (CD34, ALDH3A1, keratocan, lumican, CHST6, B3GNT7 and Col8A2) and had minimal expression of genes related to fibrosis and other lineages (vasculogenesis, adipogenesis, myogenesis, epitheliogenesis, neurogenesis and hematogenesis). Introduction of PDL spheroids into the stroma of porcine corneas resulted in extensive migration of cells inside the host stroma after 14-day organ culture. Their quiescent nature and uniform cell distribution resembled to that of mature CSKs inside the native stroma. Our results demonstrated the potential translation of PDL cells for regenerative corneal cell therapy for corneal opacities.


Asunto(s)
Células Madre Adultas/trasplante , Enfermedades de la Córnea/terapia , Ligamento Periodontal/trasplante , Medicina Regenerativa , Células Madre Adultas/citología , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Movimiento Celular/genética , Córnea/crecimiento & desarrollo , Córnea/patología , Queratocitos de la Córnea/citología , Humanos , Cresta Neural/citología , Cresta Neural/trasplante , Ligamento Periodontal/citología , Porcinos
10.
J Tissue Eng Regen Med ; 12(4): e2099-e2109, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29327452

RESUMEN

Surgical intervention is the current gold standard treatment following peripheral nerve injury. However, this approach has limitations, and full recovery of both motor and sensory modalities often remains incomplete. The development of artificial nerve grafts that either complement or replace current surgical procedures is therefore of paramount importance. An essential component of artificial grafts is biodegradable conduits and transplanted cells that provide trophic support during the regenerative process. Neural crest cells are promising support cell candidates because they are the parent population to many peripheral nervous system lineages. In this study, neural crest cells were differentiated from human embryonic stem cells. The differentiated cells exhibited typical stellate morphology and protein expression signatures that were comparable with native neural crest. Conditioned media harvested from the differentiated cells contained a range of biologically active trophic factors and was able to stimulate in vitro neurite outgrowth. Differentiated neural crest cells were seeded into a biodegradable nerve conduit, and their regeneration potential was assessed in a rat sciatic nerve injury model. A robust regeneration front was observed across the entire width of the conduit seeded with the differentiated neural crest cells. Moreover, the up-regulation of several regeneration-related genes was observed within the dorsal root ganglion and spinal cord segments harvested from transplanted animals. Our results demonstrate that the differentiated neural crest cells are biologically active and provide trophic support to stimulate peripheral nerve regeneration. Differentiated neural crest cells are therefore promising supporting cell candidates to aid in peripheral nerve repair.


Asunto(s)
Células Madre Embrionarias Humanas/metabolismo , Regeneración Nerviosa , Cresta Neural , Traumatismos de los Nervios Periféricos/terapia , Nervio Ciático , Animales , Línea Celular Tumoral , Femenino , Xenoinjertos , Humanos , Cresta Neural/metabolismo , Cresta Neural/trasplante , Ratas , Ratas Sprague-Dawley , Nervio Ciático/lesiones , Nervio Ciático/fisiología
11.
Mol Psychiatry ; 23(3): 499-508, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-27777423

RESUMEN

The enteric nervous system (ENS) is recognized as a second brain because of its complexity and its largely autonomic control of bowel function. Recent progress in studying the interactions between the ENS and the central nervous system (CNS) has implicated alterations of the gut/brain axis as a possible mechanism in the pathophysiology of autism spectrum disorders (ASDs), Parkinson's disease (PD) and other human CNS disorders, whereas the underlying mechanisms are largely unknown because of the lack of good model systems. Human induced pluripotent stem cells (hiPSCs) have the ability to proliferate indefinitely and differentiate into cells of all three germ layers, thus making iPSCs an ideal source of cells for disease modelling and cell therapy. Here, hiPSCs were induced to differentiate into neural crest stem cells (NCSCs) efficiently. When co-cultured with smooth muscle layers of ganglionic gut tissue, the NCSCs differentiated into different subtypes of mature enteric-like neurons expressing nitric oxide synthase (nNOS), vasoactive intestinal polypeptide (VIP), choline acetyltransferase (ChAT) or calretinin with typical electrophysiological characteristics of functional neurons. Furthermore, when they were transplanted into aneural or aganglionic chick, mouse or human gut tissues in ovo, in vitro or in vivo, hiPSC-derived NCSCs showed extensive migration and neural differentiation capacity, generating neurons and glial cells that expressed phenotypic markers characteristic of the enteric nervous system. Our results indicate that enteric NCSCs derived from hiPSCs supply a powerful tool for studying the pathogenesis of gastrointestinal disorders and brain/gut dysfunction and represent a potentially ideal cell source for enteric neural transplantation treatments.


Asunto(s)
Técnicas de Cocultivo/métodos , Células Madre Pluripotentes Inducidas/trasplante , Cresta Neural/fisiología , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Embrión de Pollo , Sistema Nervioso Entérico , Humanos , Ratones , Cresta Neural/trasplante , Células-Madre Neurales/fisiología , Neuronas/citología
12.
Stem Cell Reports ; 9(3): 883-896, 2017 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-28803915

RESUMEN

Acquired or congenital disruption in enteric nervous system (ENS) development or function can lead to significant mechanical dysmotility. ENS restoration through cellular transplantation may provide a cure for enteric neuropathies. We have previously generated human pluripotent stem cell (hPSC)-derived tissue-engineered small intestine (TESI) from human intestinal organoids (HIOs). However, HIO-TESI fails to develop an ENS. The purpose of our study is to restore ENS components derived exclusively from hPSCs in HIO-TESI. hPSC-derived enteric neural crest cell (ENCC) supplementation of HIO-TESI establishes submucosal and myenteric ganglia, repopulates various subclasses of neurons, and restores neuroepithelial connections and neuron-dependent contractility and relaxation in ENCC-HIO-TESI. RNA sequencing identified differentially expressed genes involved in neurogenesis, gliogenesis, gastrointestinal tract development, and differentiated epithelial cell types when ENS elements are restored during in vivo development of HIO-TESI. Our findings validate an effective approach to restoring hPSC-derived ENS components in HIO-TESI and may implicate their potential for the treatment of enteric neuropathies.


Asunto(s)
Sistema Nervioso Entérico/fisiología , Intestino Delgado/fisiología , Cresta Neural/citología , Cresta Neural/trasplante , Ingeniería de Tejidos/métodos , Transcriptoma/genética , Animales , Diferenciación Celular/genética , Línea Celular , Células Enteroendocrinas/metabolismo , Células Epiteliales/metabolismo , Perfilación de la Expresión Génica , Ontología de Genes , Humanos , Mucosa Intestinal/metabolismo , Ratones Endogámicos NOD , Ratones SCID , Neuroglía/metabolismo , Organoides/metabolismo , Células Madre Pluripotentes/metabolismo , Células Receptoras Sensoriales/metabolismo , Sinapsis/metabolismo
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.
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
15.
Artículo en Inglés | MEDLINE | ID: mdl-27380932

RESUMEN

The prospect of using neural cell replacement for the treatment of severe enteric neuropathies has seen significant progress in the last decade. The ability to harvest and transplant enteric neural crest cells (ENCCs) that functionally integrate within recipient intestine has recently been confirmed by in vivo murine studies. Although similar cells can be harvested from human fetal and postnatal gut, no studies have as yet verified their functional viability upon in vivo transplantation. We sought to determine whether ENCCs harvested from human fetal bowel are capable of engraftment and functional integration within recipient intestine following in vivo transplantation into postnatal murine colon. Enteric neural crest cells selected and harvested from fetal human gut using the neurotrophin receptor p75NTR were lentivirally labeled with either GFP or calcium-sensitive GCaMP and transplanted into the hindgut of Rag2- /γc- /C5- -immunodeficient mice at postnatal day 21. Transplanted intestines were assessed immunohistochemically for engraftment and differentiation of donor cells. Functional viability and integration with host neuromusculature was assessed using calcium imaging. Transplanted human fetal gut-derived ENCC showed engraftment within the recipient postnatal colon in 8/15 mice (53.3%). At 4 weeks posttransplantation, donor cells had spread from the site of transplantation and extended projections over distances of 1.2 ± 0.6 mm (n = 5), and differentiated into enteric nervous system (ENS) appropriate neurons and glia. These cells formed branching networks located with the myenteric plexus. Calcium transients (change in intensity F/F0 = 1.25 ± 0.03; 15 cells) were recorded in transplanted cells upon stimulation of the recipient endogenous ENS demonstrating their viability and establishment of functional connections.


Asunto(s)
Células Madre Embrionarias/trasplante , Sistema Nervioso Entérico/citología , Intestinos/citología , Intestinos/trasplante , Cresta Neural/trasplante , Células-Madre Neurales/trasplante , Animales , Células Cultivadas , Células Madre Embrionarias/fisiología , Sistema Nervioso Entérico/fisiología , Humanos , Intestinos/fisiología , Ratones , Ratones Noqueados , Cresta Neural/fisiología , Células-Madre Neurales/fisiología , Trasplante de Células Madre/métodos
16.
Dev Biol ; 419(2): 199-216, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27640086

RESUMEN

The neural crest (NC) is a remarkable transient structure generated during early vertebrate development. The neural crest progenitors have extensive migratory capacity and multipotency, harboring stem cell-like characteristics such as self-renewal. They can differentiate into a variety of cell types from craniofacial skeletal tissues to the trunk peripheral nervous system (PNS). Multiple regulators such as signaling factors, transcription factors, and migration machinery components are expressed at different stages of NC development. Gain- and loss-of-function studies in various vertebrate species revealed epistatic relationships of these molecules that could be assembled into a gene regulatory network defining the processes of NC induction, specification, migration, and differentiation. These basic developmental studies led to the subsequent establishment and molecular validation of neural crest stem cells (NCSCs) derived by various strategies. We provide here an overview of the isolation and characterization of NCSCs from embryonic, fetal, and adult tissues; the experimental strategies for the derivation of NCSCs from embryonic stem cells, induced pluripotent stem cells, and skin fibroblasts; and recent developments in the use of patient-derived NCSCs for modeling and treating neurocristopathies. We discuss future research on further refinement of the culture conditions required for the differentiation of pluripotent stem cells into axial-specific NC progenitors and their derivatives, developing non-viral approaches for the generation of induced NC cells (NCCs), and using a genomic editing approach to correct genetic mutations in patient-derived NCSCs for transplantation therapy. These future endeavors should facilitate the therapeutic applications of NCSCs in the clinical setting.


Asunto(s)
Enfermedades del Sistema Nervioso/cirugía , Cresta Neural/trasplante , Células-Madre Neurales/trasplante , Trasplante de Células Madre , Células Madre Adultas/trasplante , Animales , Técnicas de Cultivo de Célula , Linaje de la Célula , Células Cultivadas/trasplante , Desarrollo Embrionario , Transición Epitelial-Mesenquimal , Trasplante de Tejido Fetal , Predicción , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Células Madre Embrionarias Humanas/trasplante , Humanos , Células Madre Pluripotentes Inducidas/trasplante , Cresta Neural/fisiología , Neurogénesis , Especificidad de Órganos , Vertebrados/embriología
17.
Diabetes Obes Metab ; 18(2): 115-24, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26289770

RESUMEN

Despite recent advances, insulin therapy remains a treatment, not a cure, for diabetes mellitus with persistent risk of glycaemic alterations and life-threatening complications. Restoration of the endogenous ß-cell mass through regeneration or transplantation offers an attractive alternative. Unfortunately, signals that drive ß-cell regeneration remain enigmatic and ß-cell replacement therapy still faces major hurdles that prevent its widespread application. Co-transplantation of accessory non-islet cells with islet cells has been shown to improve the outcome of experimental islet transplantation. This review will highlight current travails in ß-cell therapy and focuses on the potential benefits of accessory cells for islet transplantation in diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/cirugía , Supervivencia de Injerto , Tolerancia Inmunológica , Células Secretoras de Insulina/trasplante , Trasplante de Células Madre/efectos adversos , Trasplante Heterotópico , Animales , Proliferación Celular , Separación Celular/tendencias , Células Cultivadas , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 2/inmunología , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Diabetes Mellitus Tipo 2/cirugía , Células Progenitoras Endoteliales/citología , Células Progenitoras Endoteliales/inmunología , Células Progenitoras Endoteliales/patología , Células Progenitoras Endoteliales/trasplante , Rechazo de Injerto/inmunología , Rechazo de Injerto/metabolismo , Rechazo de Injerto/prevención & control , Humanos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/metabolismo , Trasplante de Islotes Pancreáticos/efectos adversos , Trasplante de Islotes Pancreáticos/inmunología , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Trasplante de Células Madre Mesenquimatosas/tendencias , Cresta Neural/citología , Cresta Neural/inmunología , Cresta Neural/patología , Cresta Neural/trasplante , Trasplante de Células Madre/tendencias , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , Linfocitos T Reguladores/trasplante , Trasplante Autólogo/efectos adversos , Trasplante Autólogo/tendencias , Trasplante Heterotópico/efectos adversos , Trasplante Heterotópico/tendencias , Trasplante Homólogo/efectos adversos , Trasplante Homólogo/tendencias
18.
Stem Cells Transl Med ; 4(10): 1173-86, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26273065

RESUMEN

UNLABELLED: This pilot feasibility study aimed to determine the outcome of canine epidermal neural crest stem cell (cEPI-NCSC) grafts in the normal spinal cords of healthy bred-for-research dogs. This included developing novel protocols for (a) the ex vivo expansion of cEPI-NCSCs, (b) the delivery of cEPI-NCSCs into the spinal cord, and (c) the labeling of the cells and subsequent tracing of the graft in the live animal by magnetic resonance imaging. A total of four million cEPI-NCSCs were injected into the spinal cord divided in two locations. Differences in locomotion at baseline and post-treatment were evaluated by gait analysis and compared with neurological outcome and behavioral exams. Histopathological analyses of the spinal cords and cEPI-NCSC grafts were performed at 3 weeks post-transplantation. Neurological and gait parameters were minimally affected by the stem cell injection. cEPI-NCSCs survived in the canine spinal cord for the entire period of investigation and did not migrate or proliferate. Subsets of cEPI-NCSCs expressed the neural crest stem cell marker Sox10. There was no detectable expression of markers for glial cells or neurons. The tissue reaction to the cell graft was predominantly vascular in addition to a degree of reactive astrogliosis and microglial activation. In the present study, we demonstrated that cEPI-NCSC grafts survive in the spinal cords of healthy dogs without major adverse effects. They persist locally in the normal spinal cord, may promote angiogenesis and tissue remodeling, and elicit a tissue response that may be beneficial in patients with spinal cord injury. SIGNIFICANCE: It has been established that mouse and human epidermal neural crest stem cells are somatic multipotent stem cells with proved innovative potential in a mouse model of spinal cord injury (SCI) offering promise of a valid treatment for SCI. Traumatic SCI is a common neurological problem in dogs with marked similarities, clinically and pathologically, to the syndrome in people. For this reason, dogs provide a readily accessible, clinically realistic, spontaneous model for evaluation of epidermal neural crest stem cells therapeutic intervention. The results of this study are expected to give the baseline data for a future clinical trial in dogs with traumatic SCI.


Asunto(s)
Cresta Neural/trasplante , Células-Madre Neurales/trasplante , Médula Espinal/citología , Trasplante de Células Madre/métodos , Animales , Conducta Animal , Supervivencia Celular , Perros , Células Epidérmicas , Estudios de Factibilidad , Marcha , Inyecciones Espinales , Imagen por Resonancia Magnética , Ratones , Ratones Noqueados , Neurogénesis , Proyectos Piloto , Trasplante de Células Madre/efectos adversos , Teratoma , Caminata
19.
Stem Cells Transl Med ; 4(8): 905-12, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26032749

RESUMEN

Dental pulp stem cells (DPSCs) provide an exciting new avenue to study neurogenetic disorders. DPSCs are neural crest-derived cells with the ability to differentiate into numerous tissues including neurons. The therapeutic potential of stem cell-derived lines exposed to culturing ex vivo before reintroduction into patients could be limited if the cultured cells acquired tumorigenic potential. We tested whether DPSCs that spontaneously immortalized in culture acquired features of transformed cells. We analyzed immortalized DPSCs for anchorage-independent growth, genomic instability, and ability to differentiate into neurons. Finally, we tested both spontaneously immortalized and human telomerase reverse transcriptase (hTERT)-immortalized DPSC lines for the ability to form tumors in immunocompromised animals. Although we observed increased colony-forming potential in soft agar for the spontaneously immortalized and hTERT-immortalized DPSC lines relative to low-passage DPSC, no tumors were detected from any of the DPSC lines tested. We noticed some genomic instability in hTERT-immortalized DPSCs but not in the spontaneously immortalized lines tested. We determined that immortalized DPSC lines generated in our laboratory, whether spontaneously or induced, have not acquired the potential to form tumors in mice. These data suggest cultured DPSC lines that can be differentiated into neurons may be safe for future in vivo therapy for neurobiological diseases.


Asunto(s)
Pulpa Dental/trasplante , Cresta Neural/trasplante , Neuronas/citología , Trasplante de Células Madre/efectos adversos , Animales , Diferenciación Celular/genética , Transformación Celular Neoplásica , Pulpa Dental/citología , Humanos , Ratones , Telomerasa/farmacología
20.
Stem Cell Reports ; 4(5): 821-34, 2015 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-25937368

RESUMEN

Tumorigenic potential of human pluripotent stem cells (hPSCs) is an important issue in clinical applications. Despite many efforts, PSC-derived neural precursor cells (NPCs) have repeatedly induced tumors in animal models even though pluripotent cells were not detected. We found that polysialic acid-neural cell adhesion molecule (PSA-NCAM)(-) cells among the early NPCs caused tumors, whereas PSA-NCAM(+) cells were nontumorigenic. Molecular profiling, global gene analysis, and multilineage differentiation of PSA-NCAM(-) cells confirm that they are multipotent neural crest stem cells (NCSCs) that could differentiate into both ectodermal and mesodermal lineages. Transplantation of PSA-NCAM(-) cells in a gradient manner mixed with PSA-NCAM(+) cells proportionally increased mesodermal tumor formation and unwanted grafts such as PERIPHERIN(+) cells or pigmented cells in the rat brain. Therefore, we suggest that NCSCs are a critical target for tumor prevention in hPSC-derived NPCs, and removal of PSA-NCAM(-) cells eliminates the tumorigenic potential originating from NCSCs after transplantation.


Asunto(s)
Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Cresta Neural/metabolismo , Células Madre Pluripotentes/citología , Ácidos Siálicos/metabolismo , Animales , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Ectodermo/citología , Ectodermo/metabolismo , Células Madre Embrionarias Humanas/citología , Humanos , Inmunohistoquímica , Masculino , Mesodermo/citología , Mesodermo/metabolismo , Neoplasias/etiología , Neoplasias/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa/genética , Cresta Neural/citología , Cresta Neural/trasplante , Periferinas/metabolismo , Células Madre Pluripotentes/metabolismo , Ratas , Ratas Sprague-Dawley , Ácidos Siálicos/genética , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...