Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 180
Filtrar
1.
Mol Ther ; 29(4): 1439-1458, 2021 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-33309882

RESUMEN

Contrasting myelin damage through the generation of new myelinating oligodendrocytes represents a promising approach to promote functional recovery after stroke. Here, we asked whether activation of microglia and monocyte-derived macrophages affects the regenerative process sustained by G protein-coupled receptor 17 (GPR17)-expressing oligodendrocyte precursor cells (OPCs), a subpopulation of OPCs specifically reacting to ischemic injury. GPR17-iCreERT2:CAG-eGFP reporter mice were employed to trace the fate of GPR17-expressing OPCs, labeled by the green fluorescent protein (GFP), after permanent middle cerebral artery occlusion. By microglia/macrophages pharmacological depletion studies, we show that innate immune cells favor GFP+ OPC reaction and limit myelin damage early after injury, whereas they lose their pro-resolving capacity and acquire a dystrophic "senescent-like" phenotype at later stages. Intracerebral infusion of regenerative microglia-derived extracellular vesicles (EVs) restores protective microglia/macrophages functions, limiting their senescence during the post-stroke phase, and enhances the maturation of GFP+ OPCs at lesion borders, resulting in ameliorated neurological functionality. In vitro experiments show that EV-carried transmembrane tumor necrosis factor (tmTNF) mediates the pro-differentiating effects on OPCs, with future implications for regenerative therapies.


Asunto(s)
Senescencia Celular/genética , Vaina de Mielina/genética , Receptores Acoplados a Proteínas G/genética , Accidente Cerebrovascular/terapia , Animales , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Diferenciación Celular/genética , Línea Celular , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/terapia , Macrófagos/metabolismo , Macrófagos/trasplante , Masculino , Ratones , Microglía/metabolismo , Microglía/trasplante , Oligodendroglía/trasplante , Medicina Regenerativa/métodos , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/patología , Factor de Necrosis Tumoral alfa/genética
2.
Development ; 147(24)2020 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-33158923

RESUMEN

Spinal cord injury (SCI) results in loss of neurons, oligodendrocytes and myelin sheaths, all of which are not efficiently restored. The scarcity of oligodendrocytes in the lesion site impairs re-myelination of spared fibres, which leaves axons denuded, impedes signal transduction and contributes to permanent functional deficits. In contrast to mammals, zebrafish can functionally regenerate the spinal cord. Yet, little is known about oligodendroglial lineage biology and re-myelination capacity after SCI in a regeneration-permissive context. Here, we report that, in adult zebrafish, SCI results in axonal, oligodendrocyte and myelin sheath loss. We find that OPCs, the oligodendrocyte progenitor cells, survive the injury, enter a reactive state, proliferate and differentiate into oligodendrocytes. Concomitantly, the oligodendrocyte population is re-established to pre-injury levels within 2 weeks. Transcriptional profiling revealed that reactive OPCs upregulate the expression of several myelination-related genes. Interestingly, global reduction of axonal tracts and partial re-myelination, relative to pre-injury levels, persist at later stages of regeneration, yet are sufficient for functional recovery. Taken together, these findings imply that, in the zebrafish spinal cord, OPCs replace lost oligodendrocytes and, thus, re-establish myelination during regeneration.


Asunto(s)
Células Precursoras de Oligodendrocitos/citología , Remielinización/genética , Traumatismos de la Médula Espinal/genética , Médula Espinal/crecimiento & desarrollo , Animales , Modelos Animales de Enfermedad , Humanos , Células Precursoras de Oligodendrocitos/trasplante , Oligodendroglía/trasplante , Regeneración/genética , Médula Espinal/trasplante , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/terapia , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo
3.
Int J Mol Sci ; 21(19)2020 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-33008128

RESUMEN

Among the numerous candidates for cell therapy of the central nervous system (CNS), olfactory progenitors (OPs) represent an interesting alternative because they are free of ethical concerns, are easy to collect, and allow autologous transplantation. In the present study, we focused on the optimization of neuron production and maturation. It is known that plated OPs respond to various trophic factors, and we also showed that the use of Nerve Growth Factor (NGF) allowed switching from a 60/40 neuron/glia ratio to an 80/20 one. Nevertheless, in order to focus on the integration of OPs in mature neural circuits, we cocultured OPs in primary cultures obtained from the cortex and hippocampus of newborn mice. When dissociated OPs were plated, they differentiated into both glial and neuronal phenotypes, but we obtained a 1.5-fold higher viability in cortex/OP cocultures than in hippocampus/OP ones. The fate of OPs in cocultures was characterized with different markers such as BrdU, Map-2, and Synapsin, indicating a healthy integration. These results suggest that the integration of transplanted OPs might by affected by trophic factors and the environmental conditions/cell phenotypes of the host tissue. Thus, a model of coculture could provide useful information on key cell events for the use of progenitors in cell therapy.


Asunto(s)
Encéfalo/metabolismo , Neuronas/metabolismo , Corteza Olfatoria/metabolismo , Trasplante de Células Madre , Células Madre/citología , Animales , Encéfalo/citología , Encéfalo/crecimiento & desarrollo , Diferenciación Celular/genética , Linaje de la Célula/genética , Sistema Nervioso Central/metabolismo , Técnicas de Cocultivo , Humanos , Ratones , Factor de Crecimiento Nervioso/genética , Neuroglía/citología , Neuroglía/metabolismo , Neuroglía/trasplante , Neuronas/trasplante , Corteza Olfatoria/citología , Corteza Olfatoria/trasplante , Oligodendroglía/citología , Oligodendroglía/metabolismo , Oligodendroglía/trasplante , Células Madre/metabolismo
4.
Cell Stem Cell ; 25(4): 531-541.e6, 2019 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-31585094

RESUMEN

Pelizaeus-Merzbacher disease (PMD) is an X-linked leukodystrophy caused by mutations in Proteolipid Protein 1 (PLP1), encoding a major myelin protein, resulting in profound developmental delay and early lethality. Previous work showed involvement of unfolded protein response (UPR) and endoplasmic reticulum (ER) stress pathways, but poor PLP1 genotype-phenotype associations suggest additional pathogenetic mechanisms. Using induced pluripotent stem cell (iPSC) and gene-correction, we show that patient-derived oligodendrocytes can develop to the pre-myelinating stage, but subsequently undergo cell death. Mutant oligodendrocytes demonstrated key hallmarks of ferroptosis including lipid peroxidation, abnormal iron metabolism, and hypersensitivity to free iron. Iron chelation rescued mutant oligodendrocyte apoptosis, survival, and differentiationin vitro, and post-transplantation in vivo. Finally, systemic treatment of Plp1 mutant Jimpy mice with deferiprone, a small molecule iron chelator, reduced oligodendrocyte apoptosis and enabled myelin formation. Thus, oligodendrocyte iron-induced cell death and myelination is rescued by iron chelation in PMD pre-clinical models.


Asunto(s)
Deferiprona/uso terapéutico , Células Madre Pluripotentes Inducidas/fisiología , Quelantes del Hierro/uso terapéutico , Hierro/metabolismo , Proteína Proteolipídica de la Mielina/metabolismo , Oligodendroglía/fisiología , Enfermedad de Pelizaeus-Merzbacher/terapia , Animales , Diferenciación Celular , Células Cultivadas , Ferroptosis , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/trasplante , Peroxidación de Lípido , Ratones , Ratones Mutantes , Mutación/genética , Proteína Proteolipídica de la Mielina/genética , Oligodendroglía/efectos de los fármacos , Oligodendroglía/trasplante , Enfermedad de Pelizaeus-Merzbacher/genética , Enfermedad de Pelizaeus-Merzbacher/patología , Trasplante de Células Madre , Reparación del Gen Blanco
5.
CNS Neurosci Ther ; 25(10): 1075-1084, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31410988

RESUMEN

Intracerebral hemorrhage (ICH) is a cerebrovascular disorder with high mortality and disability rates. Although a lot of effort has been put in ICH, there is still no effective treatment for this devastating disease. Recent studies suggest that oligodendrocytes play an important role in brain repair after ICH and thus may be targeted for the therapies of ICH. Here in this review, we first introduce the origin, migration, proliferation, differentiation, and myelination of oligodendrocytes under physiological condition. Second, recent findings on how ICH affects oligodendrocyte biology and function are reviewed. Third, potential crosstalk between oligodendrocytes and other cells in the brain is also summarized. Last, we discuss the therapeutic potential of oligodendrocyte-based treatments in ICH. Our goal is to provide a comprehensive review on the biology and function of oligodendrocytes under both physiological and ICH conditions.


Asunto(s)
Encéfalo/metabolismo , Hemorragia Cerebral/metabolismo , Oligodendroglía/metabolismo , Encéfalo/patología , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Hemorragia Cerebral/diagnóstico , Hemorragia Cerebral/terapia , Humanos , Oligodendroglía/trasplante
6.
Mult Scler Relat Disord ; 28: 263-272, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30639828

RESUMEN

Sox2 transcription factor has been frequently used for reprograming starting cells to neural progenitor/stem cells. In vivo administration of Sox2 in the adult mouse brain reprogrammed the transduced astrocytes to neurons. In searching for adequate cell source for repairing the myelin insults, here, we studied the possible conversion of astrocytes to oligodendrocyte lineage cells by Sox2, while an extensive demyelination exists in animal brain. Lentiviral particles expressing Sox2-GFP were injected into the corpora callosa of animals fed with cuprizone diet for 12 weeks. Transduced cells were mainly astrocytes that changed their fate to oligodendrocyte lineage cells by time. For further conformation astrocytes received the vector in culture and then transplanted to the animal brains. Tracing the fate of transplanted cells showed their conversion to oligodendrocyte lineage cells. In vitro transduced cell were also maintained in the oligodendrocyte progenitor cell (OPC) induction medium. Produced OPC-like cells were positive for specific markers. This study provides a new strategy for endogenous production of myelinating cells. After optimizing the experimental conditions for safety and feasibility, this approach may contribute into future cell based therapies in patients with white matter insults as like as those with multiple sclerosis.


Asunto(s)
Astrocitos/metabolismo , Encéfalo/metabolismo , Linaje de la Célula/fisiología , Enfermedades Desmielinizantes/metabolismo , Oligodendroglía/metabolismo , Factores de Transcripción SOXB1/metabolismo , Animales , Astrocitos/patología , Astrocitos/trasplante , Encéfalo/patología , Diferenciación Celular/fisiología , Células Cultivadas , Cuprizona , Enfermedades Desmielinizantes/patología , Enfermedades Desmielinizantes/terapia , Modelos Animales de Enfermedad , Vectores Genéticos , Lentivirus/genética , Masculino , Ratones Endogámicos C57BL , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Células-Madre Neurales/trasplante , Oligodendroglía/patología , Oligodendroglía/trasplante , Regeneración/fisiología , Factores de Transcripción SOXB1/genética
7.
Macromol Biosci ; 19(2): e1800389, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30511815

RESUMEN

A recent clinical trial proves that autologous olfactory mucosal cell (OMC) transplantation improves locomotion in dogs with naturally occurring spinal injuries comparable to human lesions. However, not all dogs respond to the treatment, likely due to the transplantation procedures involving injections of cell suspensions that are associated with cell death, uneven cell distribution, and cell washout. Encapsulating cells in protective hydrogel matrices offers a tissue engineering solution to safely achieve 3D growth of viable transplant cells for implantation into injury sites, to improve regenerative outcomes. It is shown for the first time that canine OMCs (cOMCs) can be propagated with high viability in 3D collagen matrices. Further, a method to incorporate cOMCs pre-labeled with clinical-grade iron oxide nanoparticles into the constructs is described. Intraconstruct labeled cells are visualized using magnetic resonance imaging, offering substantial promise for in vivo tracking of cOMCs delivered in protective matrices.


Asunto(s)
Hidrogeles/uso terapéutico , Células-Madre Neurales/trasplante , Oligodendroglía/trasplante , Traumatismos de la Médula Espinal/terapia , Traumatismos de la Médula Espinal/veterinaria , Ingeniería de Tejidos/métodos , Animales , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Cultivadas , Colágeno/farmacología , Perros , Imagen por Resonancia Magnética , Nanopartículas de Magnetita/uso terapéutico , Modelos Animales , Mucosa Olfatoria/citología , Medicina Regenerativa/métodos , Traumatismos de la Médula Espinal/patología , Trasplante Autólogo
8.
Stem Cell Reports ; 11(6): 1433-1448, 2018 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-30472009

RESUMEN

Treatment of chronic spinal cord injury (SCI) is challenging due to cell loss, cyst formation, and the glial scar. Previously, we reported on the therapeutic potential of a neural progenitor cell (NPC) and chondroitinase ABC (ChABC) combinatorial therapy for chronic SCI. However, the source of NPCs and delivery system required for ChABC remained barriers to clinical application. Here, we investigated directly reprogrammed human NPCs biased toward an oligodendrogenic fate (oNPCs) in combination with sustained delivery of ChABC using an innovative affinity release strategy in a crosslinked methylcellulose biomaterial for the treatment of chronic SCI in an immunodeficient rat model. This combinatorial therapy increased long-term survival of oNPCs around the lesion epicenter, facilitated greater oligodendrocyte differentiation, remyelination of the spared axons by engrafted oNPCs, enhanced synaptic connectivity with anterior horn cells and neurobehavioral recovery. This combinatorial therapy is a promising strategy to regenerate the chronically injured spinal cord.


Asunto(s)
Condroitina ABC Liasa/metabolismo , Células-Madre Neurales/trasplante , Oligodendroglía/trasplante , Recuperación de la Función , Traumatismos de la Médula Espinal/fisiopatología , Animales , Células del Asta Anterior/citología , Células del Asta Anterior/metabolismo , Axones/metabolismo , Axones/ultraestructura , Diferenciación Celular , Linaje de la Célula , Supervivencia Celular , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Enfermedad Crónica , Cicatriz/patología , Reactivos de Enlaces Cruzados/química , Femenino , Humanos , Metilcelulosa/química , Actividad Motora , Neuralgia/patología , Neuralgia/fisiopatología , Plasticidad Neuronal , Ratas , Remielinización , Sinapsis/metabolismo , Sinapsis/ultraestructura
9.
Stem Cell Reports ; 10(2): 655-672, 2018 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-29337119

RESUMEN

Scarce access to primary samples and lack of efficient protocols to generate oligodendrocytes (OLs) from human pluripotent stem cells (hPSCs) are hampering our understanding of OL biology and the development of novel therapies. Here, we demonstrate that overexpression of the transcription factor SOX10 is sufficient to generate surface antigen O4-positive (O4+) and myelin basic protein-positive OLs from hPSCs in only 22 days, including from patients with multiple sclerosis or amyotrophic lateral sclerosis. The SOX10-induced O4+ population resembles primary human OLs at the transcriptome level and can myelinate neurons in vivo. Using in vitro OL-neuron co-cultures, myelination of neurons by OLs can also be demonstrated, which can be adapted to a high-throughput screening format to test the response of pro-myelinating drugs. In conclusion, we provide an approach to generate OLs in a very rapid and efficient manner, which can be used for disease modeling, drug discovery efforts, and potentially for therapeutic OL transplantation.


Asunto(s)
Diferenciación Celular/genética , Oligodendroglía/metabolismo , Células Madre Pluripotentes/metabolismo , Factores de Transcripción SOXE/genética , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/terapia , Antígenos de Superficie/genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Esclerosis Múltiple/genética , Esclerosis Múltiple/patología , Esclerosis Múltiple/terapia , Proteína Básica de Mielina/genética , Neuronas/patología , Neuronas/trasplante , Oligodendroglía/citología , Oligodendroglía/trasplante , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/trasplante , Transcriptoma/genética
10.
Mol Med Rep ; 17(1): 771-782, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29115639

RESUMEN

It has been reported that oligodendrocyte precursor cells (OPCs) may be used to treat contusive spinal cord injury (SCC), and may alter microRNA (miRNA/miR) expression following SCC in rats. However, the association between miRNA expression and the treatment of rats with SCC with OPC transplantation remain unclear. The present study transplanted OPCs into the spinal cord of rats with SCC and subsequently used the Basso, Beattie and Bresnahan (BBB) score to assess the functional recovery and pain scores. An miRNA assay was performed to detect differentially expressed miRNAs in the spinal cord of SCC rats transplanted with OPCs, compared with SCC rats transplanted with medium. Quantitative polymerase chain reaction was used to verify significantly altered miRNA expression levels. The results demonstrated that OPC transplantation was able to improve motor recovery and relieve mechanical allodynia in rats with SCC. In addition, through a miRNA assay, 45 differentially expressed miRNAs (40 upregulated miRNAs and 5 downregulated miRNAs) were detected in the spinal cord of rats in the OPC group compared with in the Medium group. Differentially expressed miRNAs were identified according to the following criteria: Fold change >2 and P<0.05. Furthermore, quantitative polymerase chain reaction was used to verify the most highly upregulated (miR­375­3p and miR­1­3p) and downregulated (miR­363­3p, miR­449a­5p and miR­3074) spinal cord miRNAs that were identified in the miRNA assay. In addition, a bioinformatics analysis of these miRNAs indicated that miR­375 and miR­1 may act primarily to inhibit cell proliferation and apoptosis via transcriptional and translational regulation, whereas miR­363, miR­449a and miR­3074 may act primarily to inhibit cell proliferation and neuronal differentiation through transcriptional regulation. These results suggested that OPC transplantation may promote functional recovery of rats with SCC, which may be associated with the expression of various miRNAs in the spinal cord, including miR­375­3p, miR­1­3p, miR­363­3p, miR­449a­5p and miR­3074.


Asunto(s)
MicroARNs/genética , Células Precursoras de Oligodendrocitos/trasplante , Oligodendroglía/trasplante , Traumatismos de la Médula Espinal/terapia , Animales , Diferenciación Celular/genética , Proliferación Celular/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Células Precursoras de Oligodendrocitos/metabolismo , Ratas , Ratas Sprague-Dawley , Recuperación de la Función , Médula Espinal/metabolismo , Médula Espinal/fisiopatología , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/fisiopatología
11.
Stem Cells Transl Med ; 6(10): 1917-1929, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28834391

RESUMEN

Cervical spinal cord injury (SCI) remains an important research focus for regenerative medicine given the potential for severe functional deficits and the current lack of treatment options to augment neurological recovery. We recently reported the preclinical safety data of a human embryonic cell-derived oligodendrocyte progenitor cell (OPC) therapy that supported initiation of a phase I clinical trial for patients with sensorimotor complete thoracic SCI. To support the clinical use of this OPC therapy for cervical injuries, we conducted preclinical efficacy and safety testing of the OPCs in a nude rat model of cervical SCI. Using the automated TreadScan system to track motor behavioral recovery, we found that OPCs significantly improved locomotor performance when administered directly into the cervical spinal cord 1 week after injury, and that this functional improvement was associated with reduced parenchymal cavitation and increased sparing of myelinated axons within the injury site. Based on large scale biodistribution and toxicology studies, we show that OPC migration is limited to the spinal cord and brainstem and did not cause any adverse clinical observations, toxicities, allodynia, or tumors. In combination with previously published efficacy and safety data, the results presented here supported initiation of a phase I/IIa clinical trial in the U.S. for patients with sensorimotor complete cervical SCI. Stem Cells Translational Medicine 2017;6:1917-1929.


Asunto(s)
Células Madre Embrionarias Humanas/citología , Células-Madre Neurales/trasplante , Oligodendroglía/trasplante , Traumatismos de la Médula Espinal/terapia , Trasplante de Células Madre/efectos adversos , Animales , Movimiento Celular , Vértebras Cervicales/lesiones , Femenino , Humanos , Células-Madre Neurales/citología , Células-Madre Neurales/fisiología , Oligodendroglía/citología , Oligodendroglía/fisiología , Ratas , Trasplante de Células Madre/métodos
12.
J Neurosurg Sci ; 61(5): 486-494, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25283064

RESUMEN

BACKGROUND: Demyelination is a common lesion in spinal cord injury, cell therapy is one of the approaches for replacing the lost oligodendrocytes. In this study, bone marrow stromal cells (BMSCs) have been transdifferentiated into oligodendrocyte-like cells (OLCs) and used in cytotherapy of contused spinal cords in rats. METHODS: The BMSCs were collected from the rat long bones, and cultured and characterized by different markers, then they were preinduced with dimethyl sulfoxide followed by retinoic acid, and then the preinduced cells were induced with combination of basic fibroblast growth factor, platelet-derived growth factor and heregulin, followed by triiodothyronine. The OLCs were transplanted in the contused spinal cords of the rats, combined with undifferentiated BMSCs. Specific markers were used in order to characterize the cells by immunohistochemistry and real-time polymerase chain reaction. The BMSCs showed typical immnuoreactivity to the markers, and the OLCs were immunostained with specific markers. RESULTS: There was an improvement in the Basso, Beattie and Bresnahan score with reduction in the cavitation in the contused rats treated with OLCs combined with BMSCs. The transplanted cells were detected in the contused spinal cord. CONCLUSIONS: The combination of the transdifferentiated BMSCs into OLCs with the undifferentiated BMSCs improved the contused spinal cord.


Asunto(s)
Transdiferenciación Celular , Trasplante de Células Madre Mesenquimatosas/métodos , Oligodendroglía/trasplante , Traumatismos de la Médula Espinal , Animales , Femenino , Células Madre Mesenquimatosas/citología , Ratas , Ratas Sprague-Dawley , Recuperación de la Función
13.
Biomaterials ; 83: 23-36, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26773663

RESUMEN

Transplantation of pluripotent stem cells and their differentiated progeny has the potential to preserve or regenerate functional pathways and improve function after central nervous system injury. However, their utility has been hampered by poor survival and the potential to form tumors. Peptide-modified biomaterials influence cell adhesion, survival and differentiation in vitro, but their effectiveness in vivo remains uncertain. We synthesized a peptide-modified, minimally invasive, injectable hydrogel comprised of hyaluronan and methylcellulose to enhance the survival and differentiation of human induced pluripotent stem cell-derived oligodendrocyte progenitor cells. Cells were transplanted subacutely after a moderate clip compression rat spinal cord injury. The hydrogel, modified with the RGD peptide and platelet-derived growth factor (PDGF-A), promoted early survival and integration of grafted cells. However, prolific teratoma formation was evident when cells were transplanted in media at longer survival times, indicating that either this cell line or the way in which it was cultured is unsuitable for human use. Interestingly, teratoma formation was attenuated when cells were transplanted in the hydrogel, where most cells differentiated to a glial phenotype. Thus, this hydrogel promoted cell survival and integration, and attenuated teratoma formation by promoting cell differentiation.


Asunto(s)
Hidrogel de Polietilenoglicol-Dimetacrilato/administración & dosificación , Células Madre Pluripotentes Inducidas/citología , Inyecciones , Oligodendroglía/citología , Traumatismos de la Médula Espinal/terapia , Teratoma/patología , Animales , Conducta Animal/efectos de los fármacos , Bovinos , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Humanos , Ácido Hialurónico/farmacología , Hidrogel de Polietilenoglicol-Dimetacrilato/farmacología , Metilcelulosa/farmacología , Oligodendroglía/trasplante , Oligopéptidos/farmacología , Factor de Crecimiento Derivado de Plaquetas/farmacología , Ratas Sprague-Dawley , Traumatismos de la Médula Espinal/patología
14.
Stem Cells ; 34(4): 984-96, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26676415

RESUMEN

Pelizaeus-Merzbacher disease (PMD) results from an X-linked misexpression of proteolipid protein 1 (PLP1). This leukodystrophy causes severe hypomyelination with progressive inflammation, leading to neurological dysfunctions and shortened life expectancy. While no cure exists for PMD, experimental cell-based therapy in the dysmyelinated shiverer model suggested that human oligodendrocyte progenitor cells (hOPCs) or human neural precursor cells (hNPCs) are promising candidates to treat myelinopathies. However, the fate and restorative advantages of human NPCs/OPCs in a relevant model of PMD has not yet been addressed. Using a model of Plp1 overexpression, resulting in demyelination with progressive inflammation, we compared side-by-side the therapeutic benefits of intracerebrally grafted hNPCs and hOPCs. Our findings reveal equal integration of the donor cells within presumptive white matter tracks. While the onset of exogenous remyelination was earlier in hOPCs-grafted mice than in hNPC-grafted mice, extended lifespan occurred only in hNPCs-grafted animals. This improved survival was correlated with reduced neuroinflammation (microglial and astrocytosis loads) and microglia polarization toward M2-like phenotype followed by remyelination. Thus modulation of neuroinflammation combined with myelin restoration is crucial to prevent PMD pathology progression and ensure successful rescue of PMD mice. These findings should help to design novel therapeutic strategies combining immunomodulation and stem/progenitor cell-based therapy for disorders associating hypomyelination with inflammation as observed in PMD.


Asunto(s)
Inmunidad Innata , Inflamación/terapia , Células-Madre Neurales/trasplante , Oligodendroglía/trasplante , Enfermedad de Pelizaeus-Merzbacher/terapia , Animales , Enfermedades Desmielinizantes/inmunología , Enfermedades Desmielinizantes/patología , Regulación del Desarrollo de la Expresión Génica , Humanos , Inmunomodulación , Inflamación/inmunología , Inflamación/patología , Ratones , Microglía/inmunología , Microglía/patología , Proteína Proteolipídica de la Mielina/biosíntesis , Vaina de Mielina/metabolismo , Células-Madre Neurales/inmunología , Oligodendroglía/inmunología , Enfermedad de Pelizaeus-Merzbacher/inmunología , Enfermedad de Pelizaeus-Merzbacher/patología , Regeneración
15.
Curr Stem Cell Res Ther ; 11(7): 569-77, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26329482

RESUMEN

There are an important number of neurological diseases where not neurons but glia are the responsible cells for the degeneration of the nervous system. In the last years, determinant roles for oligodendrocytes (OLs) have been demonstrated not only in myelin generation and maintenance but also for metabolic support of neurons. Oligodendroglial defects lead to brain degeneration in several diseases, supporting the idea that not only endogenous regeneration but also administration of exogenous OL precursors will lead to overcome functional deficits. In this review, we discuss many diseases where OLs play a crucial role, and focus on the different sources and methods to obtain oligodendroglial cells that could be used in cell therapy for myelin-related and oligodendrocyte-deficient diseases.


Asunto(s)
Enfermedades del Sistema Nervioso/terapia , Oligodendroglía/citología , Células Madre/citología , Animales , Separación Celular , Humanos , Oligodendroglía/trasplante , Fenotipo , Trasplante de Células Madre
16.
Regen Med ; 10(8): 939-58, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26345388

RESUMEN

AIM: To characterize the preclinical safety profile of a human embryonic stem cell-derived oligodendrocyte progenitor cell therapy product (AST-OPC1) in support of its use as a treatment for spinal cord injury (SCI). MATERIALS & METHODS: The phenotype and functional capacity of AST-OPC1 was characterized in vitro and in vivo. Safety and toxicology of AST-OPC1 administration was assessed in rodent models of thoracic SCI. RESULTS: These results identify AST-OPC1 as an early-stage oligodendrocyte progenitor population capable of promoting neurite outgrowth in vitro and myelination in vivo. AST-OPC1 administration did not cause any adverse clinical observations, toxicities, allodynia or tumors. CONCLUSION: These results supported initiation of a Phase I clinical trial in patients with sensorimotor complete thoracic SCI.


Asunto(s)
Células Madre Embrionarias Humanas , Oligodendroglía , Traumatismos de la Médula Espinal/terapia , Trasplante de Células Madre/métodos , Animales , Xenoinjertos , Células Madre Embrionarias Humanas/metabolismo , Células Madre Embrionarias Humanas/trasplante , Humanos , Ratones , Ratones Desnudos , Oligodendroglía/metabolismo , Oligodendroglía/trasplante , Traumatismos de la Médula Espinal/metabolismo , Trasplante de Células Madre/efectos adversos
17.
PLoS One ; 10(9): e0137211, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26352672

RESUMEN

Induced neural stem cells (iNSCs) can be directly transdifferentiated from somatic cells. One potential clinical application of the iNSCs is for nerve regeneration. However, it is unknown whether iNSCs function in disease models. We produced transdifferentiated iNSCs by conditional overexpressing Oct4, Sox2, Klf4, c-Mycin mouse embryonic fibroblasts. They expanded readily in vitro and expressed NSC mRNA profile and protein markers. These iNSCs differentiated into mature astrocytes, neurons and oligodendrocytes in vitro. Importantly, they reduced lesion size, promoted the recovery of motor and sensory function as well as metabolism status in middle cerebral artery stroke rats. These iNSCs secreted nerve growth factors, which was associated with observed protection of neurons from apoptosis. Furthermore, iNSCs migrated to and passed through the lesion in the cerebral cortex, where Tuj1+ neurons were detected. These findings have revealed the function of transdifferentiated iNSCs in vivo, and thus provide experimental evidence to support the development of personalized regenerative therapy for CNS diseases by using genetically engineered autologous somatic cells.


Asunto(s)
Transdiferenciación Celular , Corteza Cerebral/crecimiento & desarrollo , Infarto de la Arteria Cerebral Media/terapia , Regeneración Nerviosa , Células-Madre Neurales/trasplante , Animales , Astrocitos/trasplante , Diferenciación Celular/genética , Corteza Cerebral/patología , Humanos , Células Madre Pluripotentes Inducidas/trasplante , Factor 4 Similar a Kruppel , Ratones , Células-Madre Neurales/citología , Neuronas/trasplante , Oligodendroglía/trasplante , Ratas
19.
Cold Spring Harb Perspect Biol ; 7(7): a020594, 2015 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-25986556

RESUMEN

The inability of the mammalian central nervous system (CNS) to undergo spontaneous regeneration has long been regarded as a central tenet of neurobiology. However, although this is largely true of the neuronal elements of the adult mammalian CNS, save for discrete populations of granular neurons, the same is not true of its glial elements. In particular, the loss of oligodendrocytes, which results in demyelination, triggers a spontaneous and often highly efficient regenerative response, remyelination, in which new oligodendrocytes are generated and myelin sheaths are restored to denuded axons. Yet, remyelination in humans is not without limitation, and a variety of demyelinating conditions are associated with sustained and disabling myelin loss. In this review, we will review the biology of remyelination, including the cells and signals involved; describe when remyelination occurs and when and why it fails and the consequences of its failure; and discuss approaches for therapeutically enhancing remyelination in demyelinating diseases of both children and adults, both by stimulating endogenous oligodendrocyte progenitor cells and by transplanting these cells into demyelinated brain.


Asunto(s)
Vaina de Mielina/metabolismo , Regeneración Nerviosa , Enfermedades Neurodegenerativas/patología , Neuroglía/fisiología , Animales , Diferenciación Celular , Humanos , Ratones , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/terapia , Neuroglía/metabolismo , Oligodendroglía/metabolismo , Oligodendroglía/fisiología , Oligodendroglía/trasplante , Células Madre Pluripotentes/trasplante , Transducción de Señal , Trasplante de Células Madre
20.
Stem Cell Res Ther ; 6: 30, 2015 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-25888852

RESUMEN

INTRODUCTION: Globoid cell leukodystrophy (GLD) is a severe disorder of the central and peripheral nervous system caused by the absence of galactocerebrosidase (GALC) activity. Cell-based therapies are highly promising strategies for GLD. In this study, G-Olig2 mouse embryonic stem cells (ESCs) were induced into oligodendrocyte progenitor cells (OPCs) and were implanted into the brains of twitcher mice, an animal model of GLD, to explore the therapeutic potential of the cells. METHODS: The G-Olig2 ESCs were induced into OPCs by using cytokines and a multi-step differentiation procedure. Oligodendrocyte markers were detected by reverse transcription-polymerase chain reaction (RT-PCR) and immunocytochemistry. The toxicity of psychosine to OPCs was determined by a cell proliferation assay kit. The GALC level of OPCs was also examined. OPCs were labeled with Dir and transplanted into the brains of twitcher mice. The transplanted cells were detected by in-Vivo Multispectral Imaging System and real-time PCR. The physiological effects of twitcher mice were assessed. RESULTS: Oligodendrocyte markers were expressed in OPCs, and 76%±5.76% of the OPCs were enhanced green fluorescent protein (eGFP)-positive, eGFP was driven by the Olig2 promoter. The effect of psychosine on cell viability indicated that OPCs were more resistant to psychosine toxicity. The GALC level of OPCs was 10.0±1.23 nmol/hour per mg protein, which was significantly higher than other cells. Dir-labeled OPCs were injected into the forebrain of post-natal day 10 twitcher mice. The transplanted OPCs were myelin basic protein (MBP)-positive and remained along the injection tract as observed by fluorescent microscopy. The level of the Dir fluorescent signal and eGFP mRNA significantly decreased at days 10 and 20 after injection, as indicated by in-Vivo Multispectral Imaging System and real-time PCR. Because of poor cell survival and limited migration ability, there was no significant improvement in brain GALC activity, MBP level, life span, body weight, and behavioral deficits of twitcher mice. CONCLUSIONS: ESC-derived OPC transplantation was not sufficient to reverse the clinical course of GLD in twitcher mice.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Leucodistrofia de Células Globoides/terapia , Células Madre Embrionarias de Ratones/trasplante , Oligodendroglía/trasplante , Trasplante de Células Madre , Animales , Biomarcadores/metabolismo , Encéfalo/patología , Encéfalo/cirugía , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Línea Celular , Movimiento Celular , Supervivencia Celular , Modelos Animales de Enfermedad , Galactosilceramidasa/metabolismo , Ratones , Células Madre Embrionarias de Ratones/citología , Vaina de Mielina/metabolismo , Oligodendroglía/citología , Psicosina/metabolismo , Insuficiencia del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...