Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Nat Commun ; 15(1): 8570, 2024 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-39384784

RESUMEN

In Multiple Sclerosis (MS), inflammatory demyelinated lesions in the brain and spinal cord lead to neurodegeneration and progressive disability. Remyelination can restore fast saltatory conduction and neuroprotection but is inefficient in MS especially with increasing age, and is not yet treatable with therapies. Intrinsic and extrinsic inhibition of oligodendrocyte progenitor cell (OPC) function contributes to remyelination failure, and we hypothesised that the transplantation of 'improved' OPCs, genetically edited to overcome these obstacles, could improve remyelination. Here, we edit human(h) embryonic stem cell-derived OPCs to be unresponsive to a chemorepellent released from chronic MS lesions, and transplant them into rodent models of chronic lesions. Edited hOPCs display enhanced migration and remyelination compared to controls, regardless of the host age and length of time post-transplant. We show that genetic manipulation and transplantation of hOPCs overcomes the negative environment inhibiting remyelination, with translational implications for therapeutic strategies for people with progressive MS.


Asunto(s)
Esclerosis Múltiple , Células Precursoras de Oligodendrocitos , Remielinización , Animales , Remielinización/genética , Humanos , Células Precursoras de Oligodendrocitos/metabolismo , Células Precursoras de Oligodendrocitos/citología , Esclerosis Múltiple/terapia , Esclerosis Múltiple/genética , Sistemas CRISPR-Cas , Células Madre Embrionarias Humanas/metabolismo , Células Madre Embrionarias Humanas/citología , Ratas , Ratones , Edición Génica/métodos , Modelos Animales de Enfermedad , Vaina de Mielina/metabolismo , Femenino , Oligodendroglía/citología , Oligodendroglía/metabolismo , Masculino , Movimiento Celular/genética , Encefalomielitis Autoinmune Experimental/terapia , Encefalomielitis Autoinmune Experimental/genética , Diferenciación Celular
2.
Int J Mol Sci ; 22(19)2021 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-34639079

RESUMEN

Oligodendrocyte precursor cell (OPC) migration is a mechanism involved in remyelination; these cells migrate from niches in the adult CNS. However, age and disease reduce the pool of OPCs; as a result, the remyelination capacity of the CNS decreases over time. Several experimental studies have introduced OPCs to the brain via direct injection or intrathecal administration. In this study, we used the nose-to brain pathway to deliver oligodendrocyte lineage cells (human oligodendroglioma (HOG) cells), which behave similarly to OPCs in vitro. To this end, we administered GFP-labelled HOG cells intranasally to experimental animals, which were subsequently euthanised at 30 or 60 days. Our results show that the intranasal route is a viable route to the CNS and that HOG cells administered intranasally migrate preferentially to niches of OPCs (clusters created during embryonic development and adult life). Our study provides evidence, albeit limited, that HOG cells either form clusters or adhere to clusters of OPCs in the brains of experimental animals.


Asunto(s)
Encéfalo/fisiología , Enfermedades Desmielinizantes/terapia , Células Precursoras de Oligodendrocitos/citología , Oligodendroglioma/química , Remielinización , Células Madre/citología , Administración Intranasal , Animales , Encéfalo/citología , Diferenciación Celular , Células Cultivadas , Humanos
3.
Cells ; 10(8)2021 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-34440935

RESUMEN

Transplantation of oligodendrocyte precursors (OPs) is potentially therapeutic for myelin disorders but a safe and accessible cell source remains to be identified. Here we report a two-step protocol for derivation of highly enriched populations of OPs from bone marrow stromal cells of young adult rats (aMSCs). Neural progenitors among the aMSCs were expanded in non-adherent sphere-forming cultures and subsequently directed along the OP lineage with the use of glial-inducing growth factors. Immunocytochemical and flow cytometric analyses of these cells confirmed OP-like expression of Olig2, PDGFRα, NG2, and Sox10. OPs so derived formed compact myelin both in vitro, as in co-culture with purified neurons, and in vivo, following transplantation into the corpus callosum of neonatal shiverer mice. Not only did the density of myelinated axons in the corpus callosum of recipient shiverer mice reach levels comparable to those in age-matched wild-type mice, but the mean lifespan of recipient shiverer mice also far exceeded those of non-recipient shiverer mice. Our results thus promise progress in harnessing the OP-generating potential of aMSCs towards cell therapy for myelin disorders.


Asunto(s)
Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Células Precursoras de Oligodendrocitos/citología , Células Precursoras de Oligodendrocitos/metabolismo , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Proliferación Celular/fisiología , Criopreservación , Citometría de Flujo , Inmunohistoquímica , Vaina de Mielina/metabolismo , Ratas , Ratas Sprague-Dawley , Remielinización/fisiología
4.
Cells ; 10(7)2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34359956

RESUMEN

Multiple sclerosis (MS) is a neuroimmune disorder characterized by inflammation, CNS demyelination, and progressive neurodegeneration. Chronic MS patients exhibit impaired remyelination capacity, partly due to the changes that oligodendrocyte precursor cells (OPCs) undergo in response to the MS lesion environment. The cytokine tumor necrosis factor (TNF) is present in the MS-affected CNS and has been implicated in disease pathophysiology. Of the two active forms of TNF, transmembrane (tmTNF) and soluble (solTNF), tmTNF signals via TNFR2 mediating protective and reparative effects, including remyelination, whereas solTNF signals predominantly via TNFR1 promoting neurotoxicity. To better understand the mechanisms underlying repair failure in MS, we investigated the cellular responses of OPCs to inflammatory exposure and the specific role of TNFR2 signaling in their modulation. Following treatment of cultured OPCs with IFNγ, IL1ß, and TNF, we observed, by RNA sequencing, marked inflammatory and immune activation of OPCs, accompanied by metabolic changes and dysregulation of their proliferation and differentiation programming. We also established the high likelihood of cell-cell interaction between OPCs and microglia in neuroinflammatory conditions, with OPCs able to produce chemokines that can recruit and activate microglia. Importantly, we showed that these functions are exacerbated when TNFR2 is ablated. Together, our data indicate that neuroinflammation leads OPCs to shift towards an immunomodulatory phenotype while diminishing their capacity to proliferate and differentiate, thus impairing their repair function. Furthermore, we demonstrated that TNFR2 plays a key role in this process, suggesting that boosting TNFR2 activation or its downstream signals could be an effective strategy to restore OPC reparative capacity in demyelinating disease.


Asunto(s)
Diferenciación Celular/fisiología , Inmunomodulación/inmunología , Células Precursoras de Oligodendrocitos/citología , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Remielinización/fisiología , Animales , Comunicación Celular/inmunología , Inflamación/inmunología , Ratones Noqueados , Microglía/inmunología , Microglía/metabolismo , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
5.
J Cereb Blood Flow Metab ; 41(8): 2132-2133, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33969732

RESUMEN

For efficient stroke recovery, the entire neurovascular unit must be repaired. A recent study underscores this concept by highlighting the importance of cellular crosstalk for white mater remodeling. In developing brains and in brains injured by hypoxia, interactions between oligodendrocyte precursors and endothelium play an essential role for physiological and compensatory angiogenesis. Further studies are warranted to build on these emerging findings in the oligovascular niche in order to identify novel therapeutic targets for stroke and other CNS diseases.


Asunto(s)
Neovascularización Fisiológica , Células Precursoras de Oligodendrocitos/metabolismo , Animales , Comunicación Celular , Ratones , Ratones Noqueados , Células Precursoras de Oligodendrocitos/citología , Sustancia Blanca/irrigación sanguínea , Sustancia Blanca/crecimiento & desarrollo , Proteínas Wnt/deficiencia , Proteínas Wnt/genética
6.
Proc Natl Acad Sci U S A ; 118(15)2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33833053

RESUMEN

Copy number variation (CNV) at the 16p11.2 locus is associated with neuropsychiatric disorders, such as autism spectrum disorder and schizophrenia. CNVs of the 16p gene can manifest in opposing head sizes. Carriers of 16p11.2 deletion tend to have macrocephaly (or brain enlargement), while those with 16p11.2 duplication frequently have microcephaly. Increases in both gray and white matter volume have been observed in brain imaging studies in 16p11.2 deletion carriers with macrocephaly. Here, we use human induced pluripotent stem cells (hiPSCs) derived from controls and subjects with 16p11.2 deletion and 16p11.2 duplication to understand the underlying mechanisms regulating brain overgrowth. To model both gray and white matter, we differentiated patient-derived iPSCs into neural progenitor cells (NPCs) and oligodendrocyte progenitor cells (OPCs). In both NPCs and OPCs, we show that CD47 (a "don't eat me" signal) is overexpressed in the 16p11.2 deletion carriers contributing to reduced phagocytosis both in vitro and in vivo. Furthermore, 16p11.2 deletion NPCs and OPCs up-regulate cell surface expression of calreticulin (a prophagocytic "eat me" signal) and its binding sites, indicating that these cells should be phagocytosed but fail to be eliminated due to elevations in CD47. Treatment of 16p11.2 deletion NPCs and OPCs with an anti-CD47 antibody to block CD47 restores phagocytosis to control levels. While the CD47 pathway is commonly implicated in cancer progression, we document a role for CD47 in psychiatric disorders associated with brain overgrowth.


Asunto(s)
Trastorno Autístico/metabolismo , Encéfalo/metabolismo , Antígeno CD47/metabolismo , Trastornos de los Cromosomas/metabolismo , Discapacidad Intelectual/metabolismo , Adolescente , Adulto , Animales , Trastorno Autístico/patología , Encéfalo/patología , Antígeno CD47/antagonistas & inhibidores , Antígeno CD47/genética , Calreticulina/genética , Calreticulina/metabolismo , Línea Celular , Células Cultivadas , Niño , Preescolar , Deleción Cromosómica , Trastornos de los Cromosomas/patología , Cromosomas Humanos Par 16/metabolismo , Femenino , Humanos , Células Madre Pluripotentes Inducidas/citología , Discapacidad Intelectual/patología , Masculino , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Células Precursoras de Oligodendrocitos/citología , Células Precursoras de Oligodendrocitos/metabolismo
7.
Cell ; 182(3): 594-608.e11, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32679030

RESUMEN

Human cerebral cortex size and complexity has increased greatly during evolution. While increased progenitor diversity and enhanced proliferative potential play important roles in human neurogenesis and gray matter expansion, the mechanisms of human oligodendrogenesis and white matter expansion remain largely unknown. Here, we identify EGFR-expressing "Pre-OPCs" that originate from outer radial glial cells (oRGs) and undergo mitotic somal translocation (MST) during division. oRG-derived Pre-OPCs provide an additional source of human cortical oligodendrocyte precursor cells (OPCs) and define a lineage trajectory. We further show that human OPCs undergo consecutive symmetric divisions to exponentially increase the progenitor pool size. Additionally, we find that the OPC-enriched gene, PCDH15, mediates daughter cell repulsion and facilitates proliferation. These findings indicate properties of OPC derivation, proliferation, and dispersion important for human white matter expansion and myelination.


Asunto(s)
Cadherinas/metabolismo , Corteza Cerebral/citología , Células Ependimogliales/metabolismo , Neurogénesis/genética , Células Precursoras de Oligodendrocitos/metabolismo , Proteínas Relacionadas con las Cadherinas , Cadherinas/genética , Proliferación Celular/genética , Células Cultivadas , Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Células Ependimogliales/citología , Receptores ErbB/genética , Receptores ErbB/metabolismo , Células HEK293 , Humanos , Inmunohistoquímica , Células Precursoras de Oligodendrocitos/citología , ARN Interferente Pequeño , RNA-Seq , Análisis de la Célula Individual , Sustancia Blanca/citología , Sustancia Blanca/embriología , Sustancia Blanca/metabolismo
8.
Int J Mol Med ; 46(3): 1217-1224, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32582975

RESUMEN

The inflammatory cytokine interleukin (IL)­1ß has been implicated in demyelinating diseases, such as multiple sclerosis and experimental autoimmune encephalomyelitis, and brain degenerative diseases, such as Alzheimer's disease. However, the cellular and molecular mechanisms underlying the damaging effects of IL­1ß on myelination are poorly understood. Therefore, the present study was designed to investigate whether IL­1ß modifies the proliferation and differentiation of oligodendrocyte precursor cells (OPCs) through regulating the miR­202­3p/ß­catenin/glioma­associated oncogene homolog 1 (Gli1) axis. It was observed that IL­1ß significantly attenuated the proliferation and differentiation of OPCs, as evidenced by a decrease in bromodeoxyuridine incorporation and reduced percentage of myelin basic protein­positive cells among the total number of oligodendrocyte transcription factor 2­positive cells. In addition, IL­1ß markedly decreased the expression of miR­202­3p and increased the protein expression of ß­catenin and Gli1, all of which were reversed by the IL­1ß inhibitor, IL­1Ra. Treatment with the ß­catenin inhibitor XAV939, Gli1 siRNA, or miR­202­3p mimic transfection, attenuated the IL­1ß­induced suppression of OPC proliferation and differentiation. Treatment with XAV939 decreased the expression of Gli1. Transfection of miR­202­3p mimic attenuated the expression of ß­catenin and Gli1. As demonstrated by the findings of the present study, IL­1ß suppressed the proliferation and differentiation of OPCs through regulation of the miR­202­3p/ß­catenin/Gli1 axis. Therefore, the miR­202­3p/ß­catenin/Gli1 axis may be of value as a therapeutic target in multiple sclerosis.


Asunto(s)
Interleucina-1beta/metabolismo , MicroARNs/metabolismo , Células Precursoras de Oligodendrocitos/citología , Células Precursoras de Oligodendrocitos/metabolismo , Proteína con Dedos de Zinc GLI1/metabolismo , beta Catenina/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Compuestos Heterocíclicos con 3 Anillos/farmacología , Células Precursoras de Oligodendrocitos/efectos de los fármacos , beta Catenina/antagonistas & inhibidores
9.
PLoS One ; 15(5): e0233859, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32470040

RESUMEN

Mechanisms implicated in disease progression in multiple sclerosis include continued oligodendrocyte (OL)/myelin injury and failure of myelin repair. Underlying causes include metabolic stress with resultant energy deficiency. Biotin is a cofactor for carboxylases involved in ATP production that impact myelin production by promoting fatty acid synthesis. Here, we investigate the effects of high dose Biotin (MD1003) on the functional properties of post-natal rat derived oligodendrocyte progenitor cells (OPCs). A2B5 positive OPCs were assessed using an in vitro injury assay, culturing cells in either DFM (DMEM/F12+N1) or "stress media" (no glucose (NG)-DMEM), with Biotin added over a range from 2.5 to 250 µg/ml, and cell viability determined after 24 hrs. Biotin reduced the increase in OPC cell death in the NG condition. In nanofiber myelination assays, biotin increased the percentage of ensheathing cells, the number of ensheathed segments per cell, and length of ensheathed segments. In dispersed cell culture, Biotin also significantly increased ATP production, assessed using a Seahorse bio-analyzer. For most assays, the positive effects of Biotin were observed at the higher end of the dose-response analysis. We conclude that Biotin, in vitro, protects OL lineage cells from metabolic injury, enhances myelin-like ensheathment, and is associated with increased ATP production.


Asunto(s)
Adenosina Trifosfato/biosíntesis , Biotina/farmacología , Linaje de la Célula/efectos de los fármacos , Oligodendroglía/citología , Adulto , Animales , Animales Recién Nacidos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Células Precursoras de Oligodendrocitos/citología , Células Precursoras de Oligodendrocitos/efectos de los fármacos , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Ratas Sprague-Dawley
10.
Biochem Pharmacol ; 177: 113956, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32251679

RESUMEN

Oligodendrocytes are the only myelinating cells in the brain and differentiate from their progenitors (OPCs) throughout adult life. However, this process fails in demyelinating pathologies. Adenosine is emerging as an important player in OPC differentiation and we recently demonstrated that adenosine A2A receptors inhibit cell maturation by reducing voltage-dependent K+ currents. No data are available to date about the A2B receptor (A2BR) subtype. The bioactive lipid mediator sphingosine-1-phosphate (S1P) and its receptors (S1P1-5) are also crucial modulators of OPC development. An interaction between this pathway and the A2BR is reported in peripheral cells. We studied the role of A2BRs in modulating K+ currents and cell differentiation in OPC cultures and we investigated a possible interplay with S1P signaling. Our data indicate that the A2BR agonist BAY60-6583 and its new analogue P453 inhibit K+ currents in cultured OPC and the effect was prevented by the A2BR antagonist MRS1706, by K+ channel blockers and was differently modulated by the S1P analogue FTY720-P. An acute (10 min) exposure of OPCs to BAY60-6583 also increased the phosphorylated form of sphingosine kinase 1 (SphK1). A chronic (7 days) treatment with the same agonist decreased OPC differentiation whereas SphK1/2 inhibition exerted the opposite effect. Furthermore, A2BR was overexpressed during OPC differentiation, an effect prevented by the pan SphK1/2 inhibitor VPC69047. Finally, A2BR silenced cells showed increased cell maturation, decreased SphK1 expression and enhanced S1P lyase levels. We conclude that A2BRs inhibit K+ currents and cell differentiation and positively modulate S1P synthesis in cultured OPCs.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Lisofosfolípidos/farmacología , Células Precursoras de Oligodendrocitos/metabolismo , Canales de Potasio/metabolismo , Receptor de Adenosina A2B/metabolismo , Esfingosina/análogos & derivados , Aminopiridinas/farmacología , Animales , Células Cultivadas , Humanos , Células Precursoras de Oligodendrocitos/citología , Células Precursoras de Oligodendrocitos/efectos de los fármacos , Organofosfatos/farmacología , Fosforilación/efectos de los fármacos , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Purinas/farmacología , Interferencia de ARN , Ratas Wistar , Receptor de Adenosina A2B/genética , Transducción de Señal/efectos de los fármacos , Esfingosina/farmacología , Receptores de Esfingosina-1-Fosfato/metabolismo
11.
Neurosci Bull ; 36(6): 639-648, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32303914

RESUMEN

Increasing evidence suggests that a cyclic adenosine monophosphate (cAMP)-dependent intracellular signal drives the process of myelination. Yet, the signal transduction underlying the action of cAMP on central nervous system myelination remains undefined. In the present work, we sought to determine the role of EPAC (exchange protein activated by cAMP), a downstream effector of cAMP, in the development of the myelin sheath using EPAC1 and EPAC2 double-knockout (EPACdKO) mice. The results showed an age-dependent regulatory effect of EPAC1 and EPAC2 on myelin development, as their deficiency caused more myelin sheaths in postnatal early but not late adult mice. Knockout of EPAC promoted the proliferation of oligodendrocyte precursor cells and had diverse effects on myelin-related transcription factors, which in turn increased the expression of myelin-related proteins. These results indicate that EPAC proteins are negative regulators of myelination and may be promising targets for the treatment of myelin-related diseases.


Asunto(s)
Proliferación Celular , Factores de Intercambio de Guanina Nucleótido , Células Precursoras de Oligodendrocitos , Animales , AMP Cíclico , Factores de Intercambio de Guanina Nucleótido/fisiología , Ratones , Ratones Noqueados , Vaina de Mielina , Células Precursoras de Oligodendrocitos/citología
12.
Mol Neurobiol ; 57(2): 1021-1034, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31656989

RESUMEN

Neuroglial precursor cells (NPC) possess immune-modulatory properties by which they prevent immune-mediated injury in experimental autoimmune encephalomyelitis (EAE). It is unclear whether cell transplantation in a clinical-relevant setup induces ongoing therapeutic effects in a chronic-active model of progressive multiple sclerosis (MS). We examined whether human embryonic stem cell (hESC)-derived NPCs inhibit progressive EAE in Biozzi AB/H mice, manifesting with chronic-active neuroinflammation and demyelinated plaques. hESC-derived NPCs were propagated for 6-8 weeks as spheres enriched for Olig2+ cells to switch from neuronal to glial commitment and to enrich for oligodendrocyte progenitor cells. NPC were transplanted intracerebroventricularly at 30 days post-EAE induction, after the acute relapse. We evaluated effects of cell transplantation on clinical parameters, neuroinflammation, myelination, and axonal loss. Transplanted animals exhibited a significantly milder disease, reduced neuroinflammation, reduced demyelination, and reduced axonal loss as compared to control EAE mice. Toluidine-blue semi-thin staining showed a bystander neuroprotective effect of human precursor cells preventing the loss of myelinated fibers in superficial layer of the cervical dorsal funiculus. Human Olig2+ cells were detected along spinal cord meninges after 65 days of follow-up. In co-cultures in vitro, Olig2+ human precursors inhibited Concanavalin A-induced murine T cell activation and proliferation. To conclude, glial-committed human NPC induce ongoing immune-regulatory and neuroprotective effects, following transplantation into mice with a clinical-relevant model of chronic-active MS and during established disease, entering the chronic phase. These properties highlight the therapeutic potential of human NPC transplantation in chronic MS and their delivery via the cerebrospinal fluid.


Asunto(s)
Esclerosis Múltiple/inmunología , Esclerosis Múltiple/terapia , Células-Madre Neurales/trasplante , Células Precursoras de Oligodendrocitos/citología , Animales , Encefalomielitis Autoinmune Experimental/inmunología , Humanos , Ratones , Vaina de Mielina/inmunología , Neuronas/citología , Factor de Transcripción 2 de los Oligodendrocitos/metabolismo , Trasplante de Células Madre/métodos
13.
J Cell Physiol ; 235(3): 2023-2036, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31523820

RESUMEN

Oligodendrocyte precursor cells (OPCs) are considered as the main cell source for myelination in the central nervous system. Following demyelination, proliferation, migration, and differentiation capability of endogenous OPCs remarkably increase leading to remyelination in damaged areas. Despite the beneficial impacts of resident OPCs for myelin repair, the capacity of endogenous repair is low and insufficient. Therefore, several strategies have been developed to improve endogenous myelin repair. Although stem cell therapy has been introduced as a promising strategy for neurodegenerative disorders, but several limitations such as cell rejection, teratoma formation, and ethical concerns have hampered the extensive application of stem cells in clinic. In recent years, direct conversion of fully differentiated somatic cells into desired cells in the lesion area has opened a new era in regenerative medicine. In addition to direct reprogramming of somatic cells to neurons, recent evidence have also demonstrated that somatic cells, including fibroblasts and astrocytes, can be directly reprogrammed to OPC-like cells by overexpression of some specific transcription factors, microRNAs, or application of small molecules. Interestingly, induced OPCs differentiated to myelinating oligodendrocytes that could effectively ensheath the host axons. In the present review article, the current advancements in direct conversion of somatic cells towards oligodendroglial cells have been discussed both in vitro and in vivo.


Asunto(s)
Astrocitos/citología , Linaje de la Célula/fisiología , Fibroblastos/citología , Vaina de Mielina/fisiología , Células Precursoras de Oligodendrocitos/citología , Oligodendroglía/citología , Animales , Diferenciación Celular/fisiología , Humanos
14.
Cells ; 9(1)2019 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-31861635

RESUMEN

Multiple sclerosis (MS) is a demyelinating autoimmune disease of the central nervous system (CNS) mediated by autoreactive lymphocytes. The role of autoreactive lymphocytes in the CNS demyelination is well described, whereas very little is known about their role in remyelination during MS remission. In this study, we identified a new subpopulation of myelin-specific CD49d+CD154+ lymphocytes presented in the peripheral blood of MS patients during remission, that proliferated in vitro in response to myelin peptides. These lymphocytes possessed the unique ability to migrate towards maturing oligodendrocyte precursor cells (OPCs) and synthetize proinflammatory chemokines/cytokines. The co-culture of maturing OPCs with myelin-specific CD49d+CD154+ lymphocytes was characterized by the increase in proinflammatory chemokine/cytokine secretion that was not only a result of their cumulative effect of what OPCs and CD49d+CD154+ lymphocytes produced alone. Moreover, maturing OPCs exposed to exogenous myelin peptides managed to induce CD40-CD154-dependent CD49d+CD154+ lymphocyte proliferation. We confirmed, in vivo, the presence of CD49d+CD154+ cells close to maturating OPCs and remyelinating plaque during disease remission in the MS mouse model (C57Bl/6 mice immunized with MOG35-55) by immunohistochemistry. Three weeks after an acute phase of experimental autoimmune encephalomyelitis, CD49d+/CD154+ cells were found to be co-localized with O4+ cells (oligodendrocyte progenitors) in the areas of remyelination identified by myelin basic protein (MBP) labelling. These data suggested that myelin-specific CD49d+CD154+ lymphocytes present in the brain can interfere with remyelination mediated by oligodendrocytes probably as a result of establishing proinflammatory environment.


Asunto(s)
Ligando de CD40/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Integrina alfa4/metabolismo , Esclerosis Múltiple/inmunología , Vaina de Mielina/metabolismo , Adulto , Animales , Estudios de Casos y Controles , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Linfocitos/citología , Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito/efectos adversos , Células Precursoras de Oligodendrocitos/citología , Células Precursoras de Oligodendrocitos/inmunología , Fragmentos de Péptidos/efectos adversos , Remielinización
15.
Int J Mol Sci ; 20(18)2019 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-31514314

RESUMEN

Aspartate-Glutamate Carrier 1 (AGC1) deficiency is a rare neurological disease caused by mutations in the solute carrier family 25, member 12 (SLC25A12) gene, encoding for the mitochondrial aspartate-glutamate carrier isoform 1 (AGC1), a component of the malate-aspartate NADH shuttle (MAS), expressed in excitable tissues only. AGC1 deficiency patients are children showing severe hypotonia, arrested psychomotor development, seizures and global hypomyelination. While the effect of AGC1 deficiency in neurons and neuronal function has been deeply studied, little is known about oligodendrocytes and their precursors, the brain cells involved in myelination. Here we studied the effect of AGC1 down-regulation on oligodendrocyte precursor cells (OPCs), using both in vitro and in vivo mouse disease models. In the cell model, we showed that a reduced expression of AGC1 induces a deficit of OPC proliferation leading to their spontaneous and precocious differentiation into oligodendrocytes. Interestingly, this effect seems to be related to a dysregulation in the expression of trophic factors and receptors involved in OPC proliferation/differentiation, such as Platelet-Derived Growth Factor α (PDGFα) and Transforming Growth Factor ßs (TGFßs). We also confirmed the OPC reduction in vivo in AGC1-deficent mice, as well as a proliferation deficit in neurospheres from the Subventricular Zone (SVZ) of these animals, thus indicating that AGC1 reduction could affect the proliferation of different brain precursor cells. These data clearly show that AGC1 impairment alters myelination not only by acting on N-acetyl-aspartate production in neurons but also on OPC proliferation and suggest new potential therapeutic targets for the treatment of AGC1 deficiency.


Asunto(s)
Sistemas de Transporte de Aminoácidos Acídicos/deficiencia , Antiportadores/deficiencia , Mitocondrias/metabolismo , Células Precursoras de Oligodendrocitos/citología , Células Precursoras de Oligodendrocitos/metabolismo , Adenosina Trifosfato/metabolismo , Sistemas de Transporte de Aminoácidos Acídicos/metabolismo , Animales , Antiportadores/metabolismo , Diferenciación Celular , Línea Celular , Proliferación Celular , Regulación hacia Abajo , Silenciador del Gen , Lactatos/metabolismo , Ventrículos Laterales/metabolismo , Potencial de la Membrana Mitocondrial , Ratones , Neuronas/metabolismo , Factor de Crecimiento Derivado de Plaquetas , Especies Reactivas de Oxígeno/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo
16.
PLoS One ; 14(9): e0221747, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31490950

RESUMEN

Multiple sclerosis (MS) is characterized by demyelinated lesions in the central nervous system. Destruction of myelin and secondary damage to axons and neurons leads to significant disability, particularly in people with progressive MS. Accumulating evidence suggests that the potential for myelin repair exists in MS, although for unclear reasons this process fails. The cells responsible for producing myelin, the oligodendrocytes, and their progenitors, oligodendrocyte precursor cells (OPCs), have been identified at the site of lesions, even in adults. Their presence suggests the possibility that endogenous remyelination without transplantation of donor stem cells may be a mechanism for myelin repair in MS. Strategies to develop novel therapies have focused on induction of signaling pathways that stimulate OPCs to mature into myelin-producing oligodendrocytes that could then possibly remyelinate lesions. We have been investigating pharmacological approaches to enhance OPC differentiation, and have identified that the combination of two agents, triiodothyronine (T3) and quetiapine, leads to an additive effect on OPC differentiation and consequent myelin production via both overlapping and distinct signaling pathways. While the ultimate production of myelin requires cholesterol biosynthesis, we identified that quetiapine enhances gene expression in this pathway more potently than T3. Two blockers of cholesterol production, betulin and simvastatin, reduced OPC differentiation into myelin producing oligodendrocytes. Elucidating the nature of agents that lead to complementary and additive effects on oligodendrocyte differentiation and myelin production may pave the way for more efficient induction of remyelination in people with MS.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Colesterol/biosíntesis , Células Precursoras de Oligodendrocitos/citología , Células Precursoras de Oligodendrocitos/efectos de los fármacos , Fumarato de Quetiapina/farmacología , Triyodotironina/farmacología , Animales , Sinergismo Farmacológico , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Básica de Mielina/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Simvastatina/farmacología , Triterpenos/farmacología
17.
Cells ; 8(8)2019 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-31390799

RESUMEN

Remyelination in the adult brain relies on the reactivation of the Neuronal Precursor Cell (NPC) niche and differentiation into Oligodendrocyte Precursor Cells (OPCs) as well as on OPC maturation into myelinating oligodendrocytes (OLs). These two distinct phases in OL development are defined by transcriptional and morphological changes. How this differentiation program is controlled remains unclear. We used two drugs that stimulate myelin basic protein (MBP) expression (Clobetasol and Gefitinib) alone or combined with epidermal growth factor receptor (EGFR) or Retinoid X Receptor gamma (RXRγ) gene silencing to decode the receptor signaling required for OPC differentiation in myelinating OLs. Electrospun polystyrene (PS) microfibers were used as synthetic axons to study drug efficacy on fiber engagement. We show that EGFR inhibition per se stimulates MBP expression and increases Clobetasol efficacy in OPC differentiation. Consistent with this, Clobetasol and Gefitinib co-treatment, by co-regulating RXRγ, MBP and phosphatidylinositol 4,5-bisphosphate (PIP2) levels, maximizes synthetic axon engagement. Conversely, RXRγ gene silencing reduces the ability of the drugs to promote MBP expression. This work provides a view of how EGFR/ErbB inhibition controls OPC differentiation and indicates the combination of Clobetasol and Gefitinib as a potent remyelination-enhancing treatment.


Asunto(s)
Clobetasol/farmacología , Receptores ErbB/metabolismo , Gefitinib/farmacología , Proteína Básica de Mielina/metabolismo , Células Precursoras de Oligodendrocitos , Oligodendroglía , Receptor gamma X Retinoide/metabolismo , Animales , Diferenciación Celular , Línea Celular , Células Precursoras de Oligodendrocitos/citología , Células Precursoras de Oligodendrocitos/metabolismo , Oligodendroglía/citología , Oligodendroglía/metabolismo , Remielinización
18.
Sci Rep ; 9(1): 9013, 2019 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-31227736

RESUMEN

In this study, we have developed highly expandable neural stem cells (NSCs) from HESCs and iPSCs that artificially express the oligodendrocyte (OL) specific transcription factor gene Zfp488. This is enough to restrict them to an exclusive oligodendrocyte progenitor cell (OPC) fate during differentiation in vitro and in vivo. During CNS development, Zfp488 is induced during the early stages of OL generation, and then again during terminal differentiation of OLs. Interestingly, the human ortholog Znf488, crucial for OL development in human, has been recently identified to function as a dorsoventral pattering regulator in the ventral spinal cord for the generation of P1, P2/pMN, and P2 neural progenitor domains. Forced expression of Zfp488 gene in human NSCs led to the robust generation of OLs and suppression of neuronal and astrocyte fate in vitro and in vivo. Zfp488 expressing NSC derived oligodendrocytes are functional and can myelinate rat dorsal root ganglion neurons in vitro, and form myelin in Shiverer mice brain in vivo. After transplantation near a site of demyelination, Zfp488 expressing hNSCs migrated to the lesion and differentiated into premyelinating OLs. A certain fraction also homed in the subventricular zone (SVZ). Zfp488-ZsGreen1-hNSC derived OLs formed compact myelin in Shiverer mice brain seen under the electron microscope. Transplanted human neural stem cells (NSC) that have the potential to differentiate into functional oligodendrocytes in response to remyelinating signals can be a powerful therapeutic intervention for disorders where oligodendrocyte (OL) replacement is beneficial.


Asunto(s)
Diferenciación Celular , Células-Madre Neurales/citología , Neurogénesis , Neuroglía/citología , Oligodendroglía/citología , Animales , Línea Celular , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Vaina de Mielina/metabolismo , Células-Madre Neurales/metabolismo , Células-Madre Neurales/fisiología , Neuronas/citología , Neuronas/metabolismo , Células Precursoras de Oligodendrocitos/citología , Células Precursoras de Oligodendrocitos/metabolismo , Oligodendroglía/metabolismo , Oligodendroglía/fisiología , Ratas , Trasplante de Células Madre/métodos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
19.
Dev Neurosci ; 41(1-2): 79-93, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31167194

RESUMEN

Oligodendrocyte progenitor cells (OPCs) may have beneficial effects in cell replacement therapy of neurodegenerative disease owing to their unique capability to differentiate into myelinogenic oligodendrocytes (OLs) in response to extrinsic signals. Therefore, it is of significance to establish an effective differentiation methodology to generate highly pure OPCs and OLs from some easily accessible stem cell sources. To achieve this goal, in this study, we present a rapid and efficient protocol for oligodendroglial lineage differentiation from mouse neural stem cells (NSCs), rat NSCs, or mouse embryonic stem cell-derived neuroepithelial stem cells. In a defined culture medium containing Smoothened Agonist, basic fibroblast growth factor, and platelet-derived growth factor-AA, OPCs could be generated from the above stem cells over a time course of 4-6 days, achieving a cell purity as high as ∼90%. In particular, these derived OPCs showed high expandability and could further differentiate into myelin basic protein-positive OLs within 3 days or alternatively into glial fibrillary acidic protein-positive astrocytes within 7 days. Furthermore, transplantation of rodent NSC-derived OPCs into injured spinal cord indicated that it is a feasible strategy to treat spinal cord injury. Our results suggest a differentiation strategy for robust production of OPCs and OLs from rodent stem cells, which could provide an abundant OPC source for spinal cord injury.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células-Madre Neurales/citología , Células Precursoras de Oligodendrocitos/citología , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Ratones , Células Precursoras de Oligodendrocitos/trasplante , Ratas , Traumatismos de la Médula Espinal , Trasplante de Células Madre/métodos
20.
J Cell Physiol ; 234(10): 18887-18896, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30982976

RESUMEN

Oligodendrocyte (OL) loss and demyelination occur after spinal cord injury (SCI). Stimulation of remyelination through transplantation of myelinating cells may be effective in improving function. For the repair strategy to be successful, the selection of a suitable cell and maintaining cell growth when cells are injected directly to the site of injury is important. In addition to selecting the type of cell, fibrin hydrogel was used as a suitable tissue engineering scaffold for this purpose. To test the relationship between myelination and functional improvement, the human endometrial stem cells (hEnSCs) were differentiated toward oligodendrocyte progenitor cells (OPCs) using overexpression of miR-219. Adult female Wistar rats were used to induce SCI by using a compression model and were randomly assigned to the following four experimental groups: SCI, Vehicle, hEnSC, and OPC. Ten days after injury, miR-219 overexpressed hEnSC-derived OPCs encapsulated in fibrin hydrogel, as an injectable scaffold, were injected to the injury site. In this study, with a focus on promoting functional recovery after SCI, the Basso-Beattie-Bresnahan test was performed to evaluate the recovery of motor function every week for 10 weeks and the histological assay was then performed. Results showed that the rate of motor function recovery was significantly higher in OPC group compared to SCI and vehicle groups but no marked differences were found between OPC and hEnSC groups, although, the rate of myelination in the OPC group was significantly higher than the other groups. These results demonstrated that remyelination was not the cause of recovery of motor function.


Asunto(s)
MicroARNs/biosíntesis , Regeneración Nerviosa/fisiología , Células Precursoras de Oligodendrocitos/citología , Traumatismos de la Médula Espinal/terapia , Trasplante de Células Madre/métodos , Células Madre/citología , Animales , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Cultivadas , Endometrio/citología , Femenino , Fibrina/uso terapéutico , Humanos , Hidrogeles/uso terapéutico , MicroARNs/genética , Ratas , Ratas Wistar , Remielinización/fisiología , Traumatismos de la Médula Espinal/fisiopatología , Ingeniería de Tejidos/métodos , Andamios del Tejido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA