Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 300(1): 105487, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37995941

RESUMEN

Oligodendrocyte precursor cells are present in the adult central nervous system, and their impaired ability to differentiate into myelinating oligodendrocytes can lead to demyelination in patients with multiple sclerosis, accompanied by neurological deficits and cognitive impairment. Exosomes, small vesicles released by cells, are known to facilitate intercellular communication by carrying bioactive molecules. In this study, we utilized exosomes derived from human umbilical cord mesenchymal stem cells (HUMSCs-Exos). We performed sequencing and bioinformatics analysis of exosome-treated cells to demonstrate that HUMSCs-Exos can stimulate myelin gene expression in oigodendrocyte precursor cells. Functional investigations revealed that HUMSCs-Exos activate the Pi3k/Akt pathway and regulate the Tbr1/Wnt signaling molecules through the transfer of miR-23a-3p, promoting oligodendrocytes differentiation and enhancing the expression of myelin-related proteins. In an experimental autoimmune encephalomyelitis model, treatment with HUMSCs-Exos significantly improved neurological function and facilitated remyelination. This study provides cellular and molecular insights into the use of cell-free exosome therapy for central nervous system demyelination associated with multiple sclerosis, demonstrating its great potential for treating demyelinating and neurodegenerative diseases.


Asunto(s)
Exosomas , Células Madre Mesenquimatosas , MicroARNs , Esclerosis Múltiple , Remielinización , Adulto , Humanos , Diferenciación Celular/genética , Exosomas/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , MicroARNs/metabolismo , MicroARNs/farmacología , MicroARNs/uso terapéutico , Esclerosis Múltiple/genética , Esclerosis Múltiple/terapia , Esclerosis Múltiple/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Remielinización/efectos de los fármacos , Remielinización/genética , Cordón Umbilical/citología , Cordón Umbilical/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteínas de Dominio T Box/metabolismo , Modelos Animales de Enfermedad , Células Cultivadas
2.
Int J Mol Sci ; 23(23)2022 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-36499195

RESUMEN

Demyelinating disorders show impaired remyelination due to failure in the differentiation of oligodendrocyte progenitor cells (OPCs) into mature myelin-forming oligodendrocytes, a process driven by microglia-OPC crosstalk. Through conducting a transcriptomic analysis of microarray studies on the demyelination-remyelination cuprizone model and using human samples of multiple sclerosis (MS), we identified molecules involved in this crosstalk. Differentially expressed genes (DEGs) of specific regions/cell types were detected in GEO transcriptomic raw data after cuprizone treatment and in MS samples, followed by functional analysis with GO terms and WikiPathways. Additionally, microglia-OPC crosstalk between microglia ligands, OPC receptors and target genes was examined with the NicheNet model. We identified 108 and 166 DEGs in the demyelinated corpus callosum (CC) at 2 and 4 weeks of cuprizone treatment; 427 and 355 DEGs in the remyelinated (4 weeks of cuprizone treatment + 14 days of normal diet) compared to 2- and 4-week demyelinated CC; 252 DEGs in MS samples and 2730 and 12 DEGs in OPC and microglia of 4-week demyelinated CC. At this time point, we found 95 common DEGs in the CC and OPCs, and one common DEG in microglia and OPCs, mostly associated with myelin and lipid metabolism. Crosstalk analysis identified 47 microglia ligands, 43 OPC receptors and 115 OPC target genes, all differentially expressed in cuprizone-treated samples and associated with myelination. Our differential expression pipeline identified demyelination/remyelination transcriptomic biomarkers in studies using diverse platforms and cell types/tissues. Cellular crosstalk analysis yielded novel markers of microglia ligands, OPC receptors and target genes.


Asunto(s)
Enfermedades Desmielinizantes , Esclerosis Múltiple , Células Precursoras de Oligodendrocitos , Remielinización , Ratones , Animales , Humanos , Células Precursoras de Oligodendrocitos/metabolismo , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/genética , Enfermedades Desmielinizantes/metabolismo , Ratones Endogámicos C57BL , Remielinización/genética , Cuprizona/toxicidad , Oligodendroglía/metabolismo , Vaina de Mielina/genética , Vaina de Mielina/metabolismo , Diferenciación Celular/genética , Microglía/metabolismo , Esclerosis Múltiple/genética , Esclerosis Múltiple/metabolismo , Modelos Animales de Enfermedad
3.
Cell Rep ; 37(4): 109889, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34706227

RESUMEN

Astrocyte-derived cholesterol supports brain cells under physiological conditions. However, in demyelinating lesions, astrocytes downregulate cholesterol synthesis, and the cholesterol that is essential for remyelination has to originate from other cellular sources. Here, we show that repair following acute versus chronic demyelination involves distinct processes. In particular, in chronic myelin disease, when recycling of lipids is often defective, de novo neuronal cholesterol synthesis is critical for regeneration. By gene expression profiling, genetic loss-of-function experiments, and comprehensive phenotyping, we provide evidence that neurons increase cholesterol synthesis in chronic myelin disease models and in patients with multiple sclerosis (MS). In mouse models, neuronal cholesterol facilitates remyelination specifically by triggering oligodendrocyte precursor cell proliferation. Our data contribute to the understanding of disease progression and have implications for therapeutic strategies in patients with MS.


Asunto(s)
Colesterol , Esclerosis Múltiple , Vaina de Mielina , Células Precursoras de Oligodendrocitos/metabolismo , Remielinización/genética , Animales , Colesterol/biosíntesis , Colesterol/genética , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Noqueados , Esclerosis Múltiple/genética , Esclerosis Múltiple/metabolismo , Vaina de Mielina/genética , Vaina de Mielina/metabolismo
4.
Cell Mol Life Sci ; 78(19-20): 6631-6644, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34482420

RESUMEN

The differentiation of oligodendrocyte precursor cells (OPCs) into myelinating oligodendrocytes is the prerequisite for remyelination in demyelinated disorders such as multiple sclerosis (MS). Epigenetic mechanisms, such as DNA methylation, have been suggested to control the intricate network of transcription factors involved in OPC differentiation. Yet, the exact mechanism remains undisclosed. Here, we are the first to identify the DNA-binding protein inhibitors, Id2 and Id4, as targets of DNA methylation during OPC differentiation. Using state-of-the-art epigenetic editing via CRISPR/dCas9-DNMT3a, we confirm that targeted methylation of Id2/Id4 drives OPC differentiation. Moreover, we show that in the pathological context of MS, methylation and gene expression levels of both ID2 and ID4 are altered compared to control human brain samples. We conclude that DNA methylation is crucial to suppress ID2 and ID4 during OPC differentiation, a process that appears to be dysregulated during MS. Our data do not only reveal new insights into oligodendrocyte biology, but could also lead to a better understanding of CNS myelin disorders.


Asunto(s)
Diferenciación Celular/genética , Metilación de ADN/genética , Regulación de la Expresión Génica/genética , Expresión Génica/genética , Proteína 2 Inhibidora de la Diferenciación/genética , Proteínas Inhibidoras de la Diferenciación/genética , Factores de Transcripción/genética , Animales , Células Cultivadas , Epigénesis Genética/genética , Ratones , Vaina de Mielina/genética , Células Precursoras de Oligodendrocitos/fisiología , Remielinización/genética
5.
Neuron ; 109(19): 3104-3118.e6, 2021 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-34390652

RESUMEN

Oligodendrocyte (OL) maturation arrest in human white matter injury contributes significantly to the failure of endogenous remyelination in multiple sclerosis (MS) and newborn brain injuries such as hypoxic ischemic encephalopathy (HIE) that cause cerebral palsy. In this study, we identify an oligodendroglial-intrinsic factor that controls OL maturation specifically in the setting of injury. We find a requirement for the ring finger protein Rnf43 not in normal development but in neonatal hypoxic injury and remyelination in the adult mammalian CNS. Rnf43, but not the related Znrf3, is potently activated by Wnt signaling in OL progenitor cells (OPCs) and marks activated OPCs in human MS and HIE. Rnf43 is required in an injury-specific context, and it promotes OPC differentiation through negative regulation of Wnt signal strength in OPCs at the level of Fzd1 receptor presentation on the cell surface. Inhibition of Fzd1 using UM206 promotes remyelination following ex vivo and in vivo demyelinating injury.


Asunto(s)
Lesiones Encefálicas/genética , Lesiones Encefálicas/patología , Oligodendroglía/patología , Ubiquitina-Proteína Ligasas/genética , Animales , Lesiones Encefálicas/metabolismo , Enfermedades Desmielinizantes/genética , Receptores Frizzled/efectos de los fármacos , Receptores Frizzled/genética , Humanos , Ratones , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/fisiología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Remielinización/efectos de los fármacos , Remielinización/genética , Células Madre/metabolismo , Células Madre/patología , Sustancia Blanca/metabolismo , Sustancia Blanca/patología , Vía de Señalización Wnt
6.
Neurobiol Dis ; 159: 105494, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34464706

RESUMEN

The gene GPNMB is known to play roles in phagocytosis and tissue repair, and is upregulated in microglia in many mouse models of neurodegenerative disease as well as in human patients. Nearby genomic variants are associated with both elevated Parkinson's disease (PD) risk and higher expression of this gene, suggesting that inhibiting GPNMB activity might be protective in Parkinson's disease. We tested this hypothesis in three different mouse models of neurological diseases: a remyelination model and two models of alpha-synuclein pathology. We found that Gpnmb deletion had no effect on histological, cellular, behavioral, neurochemical or gene expression phenotypes in any of these models. These data suggest that Gpnmb does not play a major role in the development of pathology or functional defects in these models and that further work is necessary to study its role in the development or progression of Parkinson's disease.


Asunto(s)
Proteínas del Ojo/genética , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Enfermedad de Parkinson/metabolismo , Remielinización/genética , Sustancia Negra/metabolismo , Sinucleinopatías/genética , Anciano , Anciano de 80 o más Años , Animales , Encéfalo/metabolismo , Encéfalo/patología , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Enfermedad de Parkinson/patología , Sustancia Negra/patología , Sinucleinopatías/metabolismo , Sinucleinopatías/patología
7.
Nat Commun ; 12(1): 3359, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-34099715

RESUMEN

The mechanisms regulating myelin repair in the adult central nervous system (CNS) are unclear. Here, we identify DNA hydroxymethylation, catalyzed by the Ten-Eleven-Translocation (TET) enzyme TET1, as necessary for myelin repair in young adults and defective in old mice. Constitutive and inducible oligodendrocyte lineage-specific ablation of Tet1 (but not of Tet2), recapitulate this age-related decline in repair of demyelinated lesions. DNA hydroxymethylation and transcriptomic analyses identify TET1-target in adult oligodendrocytes, as genes regulating neuro-glial communication, including the solute carrier (Slc) gene family. Among them, we show that the expression levels of the Na+/K+/Cl- transporter, SLC12A2, are higher in Tet1 overexpressing cells and lower in old or Tet1 knockout. Both aged mice and Tet1 mutants also present inefficient myelin repair and axo-myelinic swellings. Zebrafish mutants for slc12a2b also display swellings of CNS myelinated axons. Our findings suggest that TET1 is required for adult myelin repair and regulation of the axon-myelin interface.


Asunto(s)
Metilación de ADN , Proteínas de Unión al ADN/genética , Perfilación de la Expresión Génica/métodos , Vaina de Mielina/genética , Proteínas Proto-Oncogénicas/genética , Remielinización/genética , Animales , Animales Modificados Genéticamente , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Ratones Noqueados , Ratones Transgénicos , Mutación , Vaina de Mielina/metabolismo , Oligodendroglía/citología , Oligodendroglía/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , RNA-Seq/métodos , Miembro 2 de la Familia de Transportadores de Soluto 12/genética , Miembro 2 de la Familia de Transportadores de Soluto 12/metabolismo , Pez Cebra/genética
8.
Cells ; 10(4)2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33924474

RESUMEN

Multiple sclerosis (MS) is a chronic inflammatory and neurodegenerative disease of the central nervous system (CNS) affecting more than two million people worldwide. In MS, oligodendrocytes and myelin sheaths are destroyed by autoimmune-mediated inflammation, while remyelination is impaired. Recent investigations of post-mortem tissue suggest that Fibroblast growth factor (FGF) signaling may regulate inflammation and myelination in MS. FGF2 expression seems to correlate positively with macrophages/microglia and negatively with myelination; FGF1 was suggested to promote remyelination. In myelin oligodendrocyte glycoprotein (MOG)35-55-induced experimental autoimmune encephalomyelitis (EAE), systemic deletion of FGF2 suggested that FGF2 may promote remyelination. Specific deletion of FGF receptors (FGFRs) in oligodendrocytes in this EAE model resulted in a decrease of lymphocyte and macrophage/microglia infiltration as well as myelin and axon degeneration. These effects were mediated by ERK/Akt phosphorylation, a brain-derived neurotrophic factor, and downregulation of inhibitors of remyelination. In the first part of this review, the most important pharmacotherapeutic principles for MS will be illustrated, and then we will review recent advances made on FGF signaling in MS. Thus, we will suggest application of FGFR inhibitors, which are currently used in Phase II and III cancer trials, as a therapeutic option to reduce inflammation and induce remyelination in EAE and eventually MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/genética , Factor 2 de Crecimiento de Fibroblastos/genética , Microglía/inmunología , Esclerosis Múltiple/genética , Vaina de Mielina/inmunología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/inmunología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Factor 2 de Crecimiento de Fibroblastos/deficiencia , Regulación de la Expresión Génica , Humanos , Factores Inmunológicos/uso terapéutico , Ratones Noqueados , Microglía/efectos de los fármacos , Microglía/patología , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/patología , Glicoproteína Mielina-Oligodendrócito/administración & dosificación , Oligodendroglía/efectos de los fármacos , Oligodendroglía/inmunología , Oligodendroglía/patología , Fragmentos de Péptidos/administración & dosificación , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/inmunología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/inmunología , Remielinización/efectos de los fármacos , Remielinización/genética , Remielinización/inmunología , Transducción de Señal
9.
Nat Commun ; 12(1): 1923, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33772011

RESUMEN

Chronic demyelination in the human CNS is characterized by an inhibitory microenvironment that impairs recruitment and differentiation of oligodendrocyte progenitor cells (OPCs) leading to failed remyelination and axonal atrophy. By network-based transcriptomics, we identified sulfatase 2 (Sulf2) mRNA in activated human primary OPCs. Sulf2, an extracellular endosulfatase, modulates the signaling microenvironment by editing the pattern of sulfation on heparan sulfate proteoglycans. We found that Sulf2 was increased in demyelinating lesions in multiple sclerosis and was actively secreted by human OPCs. In experimental demyelination, elevated OPC Sulf1/2 expression directly impaired progenitor recruitment and subsequent generation of oligodendrocytes thereby limiting remyelination. Sulf1/2 potentiates the inhibitory microenvironment by promoting BMP and WNT signaling in OPCs. Importantly, pharmacological sulfatase inhibition using PI-88 accelerated oligodendrocyte recruitment and remyelination by blocking OPC-expressed sulfatases. Our findings define an important inhibitory role of Sulf1/2 and highlight the potential for modulation of the heparanome in the treatment of chronic demyelinating disease.


Asunto(s)
Diferenciación Celular/genética , Microambiente Celular/genética , Enfermedades Desmielinizantes/genética , Perfilación de la Expresión Génica/métodos , Células Precursoras de Oligodendrocitos/metabolismo , Remielinización/genética , Animales , Axones/metabolismo , Células Cultivadas , Enfermedades Desmielinizantes/metabolismo , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos , Esclerosis Múltiple/genética , Esclerosis Múltiple/metabolismo , Células Precursoras de Oligodendrocitos/citología , Sulfatasas/genética , Sulfatasas/metabolismo , Sulfotransferasas/genética , Sulfotransferasas/metabolismo
10.
Elife ; 102021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33752802

RESUMEN

The inflammatory environment of demyelinated lesions in multiple sclerosis (MS) patients contributes to remyelination failure. Inflammation activates a cytoprotective pathway, the integrated stress response (ISR), but it remains unclear whether enhancing the ISR can improve remyelination in an inflammatory environment. To examine this possibility, the remyelination stage of experimental autoimmune encephalomyelitis (EAE), as well as a mouse model that incorporates cuprizone-induced demyelination along with CNS delivery of the proinflammatory cytokine IFN-γ were used here. We demonstrate that either genetic or pharmacological ISR enhancement significantly increased the number of remyelinating oligodendrocytes and remyelinated axons in the inflammatory lesions. Moreover, the combined treatment of the ISR modulator Sephin1 with the oligodendrocyte differentiation enhancing reagent bazedoxifene increased myelin thickness of remyelinated axons to pre-lesion levels. Taken together, our findings indicate that prolonging the ISR protects remyelinating oligodendrocytes and promotes remyelination in the presence of inflammation, suggesting that ISR enhancement may provide reparative benefit to MS patients.


Asunto(s)
Sistema Nervioso Central/inmunología , Cuprizona/efectos adversos , Enfermedades Desmielinizantes/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Remielinización/fisiología , Animales , Axones/inmunología , Enfermedades Desmielinizantes/inducido químicamente , Modelos Animales de Enfermedad , Femenino , Inflamación/genética , Inflamación/inmunología , Interferón gamma/genética , Interferón gamma/metabolismo , Masculino , Ratones , Oligodendroglía/inmunología , Remielinización/genética
11.
Sci Rep ; 11(1): 4132, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33603109

RESUMEN

To identify markers in the CSF of multiple sclerosis (MS) subtypes, we used a two-step proteomic approach: (i) Discovery proteomics compared 169 pooled CSF from MS subtypes and inflammatory/degenerative CNS diseases (NMO spectrum and Alzheimer disease) and healthy controls. (ii) Next, 299 proteins selected by comprehensive statistics were quantified in 170 individual CSF samples. (iii) Genes of the identified proteins were also screened among transcripts in 73 MS brain lesions compared to 25 control brains. F-test based feature selection resulted in 8 proteins differentiating the MS subtypes, and secondary progressive (SP)MS was the most different also from controls. Genes of 7 out these 8 proteins were present in MS brain lesions: GOLM was significantly differentially expressed in active, chronic active, inactive and remyelinating lesions, FRZB in active and chronic active lesions, and SELENBP1 in inactive lesions. Volcano maps of normalized proteins in the different disease groups also indicated the highest amount of altered proteins in SPMS. Apolipoprotein C-I, apolipoprotein A-II, augurin, receptor-type tyrosine-protein phosphatase gamma, and trypsin-1 were upregulated in the CSF of MS subtypes compared to controls. This CSF profile and associated brain lesion spectrum highlight non-inflammatory mechanisms in differentiating CNS diseases and MS subtypes and the uniqueness of SPMS.


Asunto(s)
Encéfalo/metabolismo , Líquido Cefalorraquídeo/metabolismo , Esclerosis Múltiple Crónica Progresiva/líquido cefalorraquídeo , Esclerosis Múltiple Crónica Progresiva/metabolismo , Proteoma/genética , Proteoma/metabolismo , Transcriptoma/genética , Adulto , Biomarcadores/líquido cefalorraquídeo , Femenino , Humanos , Masculino , Esclerosis Múltiple Crónica Progresiva/genética , Proteómica/métodos , Remielinización/genética , Proteínas de Unión al Selenio/genética , Proteínas de Unión al Selenio/metabolismo
12.
PLoS One ; 16(1): e0245944, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33481951

RESUMEN

The cellular prion protein (PrP) is essential to the long-term maintenance of myelin sheaths in peripheral nerves. PrP activates the adhesion G-protein coupled receptor Adgrg6 on Schwann cells and initiates a pro-myelination cascade of molecular signals. Because Adgrg6 is crucial for peripheral myelin development and regeneration after nerve injury, we investigated the role of PrP in peripheral nerve repair. We performed experimental sciatic nerve crush injuries in co-isogenic wild-type and PrP-deficient mice, and examined peripheral nerve repair processes. Generation of repair Schwann cells, macrophage recruitment and remyelination were similar in PrP-deficient and wild-type mice. We conclude that PrP is dispensable for sciatic nerve de- and remyelination after crush injury. Adgrg6 may sustain its function in peripheral nerve repair independently of its activation by PrP.


Asunto(s)
Regeneración Nerviosa/fisiología , Traumatismos de los Nervios Periféricos/metabolismo , Proteínas Priónicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Remielinización/genética , Células de Schwann/metabolismo , Nervio Ciático/metabolismo , Animales , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Vaina de Mielina/metabolismo , Compresión Nerviosa , Traumatismos de los Nervios Periféricos/genética , Proteínas Priónicas/genética , Receptores Acoplados a Proteínas G/genética , Nervio Ciático/lesiones
13.
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
14.
Science ; 370(6512)2020 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-33004487

RESUMEN

Injuries to the central nervous system (CNS) are inefficiently repaired. Resident neural stem cells manifest a limited contribution to cell replacement. We have uncovered a latent potential in neural stem cells to replace large numbers of lost oligodendrocytes in the injured mouse spinal cord. Integrating multimodal single-cell analysis, we found that neural stem cells are in a permissive chromatin state that enables the unfolding of a normally latent gene expression program for oligodendrogenesis after injury. Ectopic expression of the transcription factor OLIG2 unveiled abundant stem cell-derived oligodendrogenesis, which followed the natural progression of oligodendrocyte differentiation, contributed to axon remyelination, and stimulated functional recovery of axon conduction. Recruitment of resident stem cells may thus serve as an alternative to cell transplantation after CNS injury.


Asunto(s)
Células-Madre Neurales/fisiología , Neurogénesis/fisiología , Oligodendroglía/fisiología , Regeneración de la Medula Espinal/fisiología , Animales , Astrocitos/fisiología , Axones/fisiología , Linaje de la Célula , Epéndimo/citología , Epéndimo/metabolismo , Ratones , Ratones Endogámicos C57BL , Neurogénesis/genética , Factor de Transcripción 2 de los Oligodendrocitos/metabolismo , Oligodendroglía/citología , Recuperación de la Función/genética , Recuperación de la Función/fisiología , Remielinización/genética , Remielinización/fisiología , Análisis de la Célula Individual , Traumatismos de la Médula Espinal/fisiopatología , Regeneración de la Medula Espinal/genética
15.
Proc Natl Acad Sci U S A ; 117(39): 24464-24474, 2020 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-32929007

RESUMEN

Microglia are considered both pathogenic and protective during recovery from demyelination, but their precise role remains ill defined. Here, using an inhibitor of colony stimulating factor 1 receptor (CSF1R), PLX5622, and mice infected with a neurotropic coronavirus (mouse hepatitis virus [MHV], strain JHMV), we show that depletion of microglia during the time of JHMV clearance resulted in impaired myelin repair and prolonged clinical disease without affecting the kinetics of virus clearance. Microglia were required only during the early stages of remyelination. Notably, large deposits of extracellular vesiculated myelin and cellular debris were detected in the spinal cords of PLX5622-treated and not control mice, which correlated with decreased numbers of oligodendrocytes in demyelinating lesions in drug-treated mice. Furthermore, gene expression analyses demonstrated differential expression of genes involved in myelin debris clearance, lipid and cholesterol recycling, and promotion of oligodendrocyte function. The results also demonstrate that microglial functions affected by depletion could not be compensated by infiltrating macrophages. Together, these results demonstrate that microglia play key roles in debris clearance and in the initiation of remyelination following infection with a neurotropic coronavirus but are not necessary during later stages of remyelination.


Asunto(s)
Infecciones por Coronavirus/patología , Enfermedades Desmielinizantes/patología , Microglía/patología , Remielinización , Animales , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Enfermedades Desmielinizantes/inmunología , Enfermedades Desmielinizantes/virología , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Inmunidad Celular/efectos de los fármacos , Inflamación , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/metabolismo , Virus de la Hepatitis Murina/efectos de los fármacos , Virus de la Hepatitis Murina/fisiología , Vaina de Mielina/metabolismo , Vaina de Mielina/patología , Oligodendroglía/patología , Compuestos Orgánicos/administración & dosificación , Compuestos Orgánicos/efectos adversos , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Remielinización/genética , Médula Espinal/inmunología , Médula Espinal/patología
16.
Nat Commun ; 11(1): 4071, 2020 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-32792491

RESUMEN

Arrest of oligodendrocyte (OL) differentiation and remyelination following myelin damage in multiple sclerosis (MS) is associated with neurodegeneration and clinical worsening. We show that Glutathione S-transferase 4α (Gsta4) is highly expressed during adult OL differentiation and that Gsta4 loss impairs differentiation into myelinating OLs in vitro. In addition, we identify Gsta4 as a target of both dimethyl fumarate, an existing MS therapy, and clemastine fumarate, a candidate remyelinating agent in MS. Overexpression of Gsta4 reduces expression of Fas and activity of the mitochondria-associated Casp8-Bid-axis in adult oligodendrocyte precursor cells, leading to improved OL survival during differentiation. The Gsta4 effect on apoptosis during adult OL differentiation was corroborated in vivo in both lysolecithin-induced demyelination and experimental autoimmune encephalomyelitis models, where Casp8 activity was reduced in Gsta4-overexpressing OLs. Our results identify Gsta4 as an intrinsic regulator of OL differentiation, survival and remyelination, as well as a potential target for future reparative MS therapies.


Asunto(s)
Glutatión Transferasa/metabolismo , Oligodendroglía/citología , Oligodendroglía/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Caspasa 8/genética , Caspasa 8/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Glutatión Transferasa/genética , Homeostasis/genética , Homeostasis/fisiología , Inmunohistoquímica , Masculino , Microglía/citología , Microglía/metabolismo , Microscopía Electrónica de Transmisión , Mitocondrias/metabolismo , Esclerosis Múltiple/genética , Esclerosis Múltiple/metabolismo , Fagocitosis/genética , Fagocitosis/fisiología , Procesamiento Proteico-Postraduccional , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Remielinización/genética , Remielinización/fisiología
17.
Proc Natl Acad Sci U S A ; 117(30): 18018-18028, 2020 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-32651278

RESUMEN

CCN3 is a matricellular protein that promotes oligodendrocyte progenitor cell differentiation and myelination in vitro and ex vivo. CCN3 is therefore a candidate of interest in central nervous system (CNS) myelination and remyelination, and we sought to investigate the expression and role of CCN3 during these processes. We found CCN3 to be expressed predominantly by neurons in distinct areas of the CNS, primarily the cerebral cortex, hippocampus, amygdala, suprachiasmatic nuclei, anterior olfactory nuclei, and spinal cord gray matter. CCN3 was transiently up-regulated following demyelination in the brain of cuprizone-fed mice and spinal cord lesions of mice injected with lysolecithin. However, CCN3-/- mice did not exhibit significantly different numbers of oligodendroglia or differentiated oligodendrocytes in the healthy or remyelinating CNS, compared to WT controls. These results suggest that despite robust and dynamic expression in the CNS, CCN3 is not required for efficient myelination or remyelination in the murine CNS in vivo.


Asunto(s)
Sistema Nervioso Central/metabolismo , Enfermedades Desmielinizantes/etiología , Regulación de la Expresión Génica , Proteína Hiperexpresada del Nefroblastoma/genética , Remielinización/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Ratones , Vaina de Mielina/metabolismo , Proteína Hiperexpresada del Nefroblastoma/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Oligodendroglía/metabolismo , Médula Espinal/metabolismo , Médula Espinal/patología
18.
Nat Commun ; 11(1): 3420, 2020 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-32647127

RESUMEN

Remyelination of the peripheral and central nervous systems (PNS and CNS, respectively) is a prerequisite for functional recovery after lesion. However, this process is not always optimal and becomes inefficient in the course of multiple sclerosis. Here we show that, when acetylated, eukaryotic elongation factor 1A1 (eEF1A1) negatively regulates PNS and CNS remyelination. Acetylated eEF1A1 (Ac-eEF1A1) translocates into the nucleus of myelinating cells where it binds to Sox10, a key transcription factor for PNS and CNS myelination and remyelination, to drag Sox10 out of the nucleus. We show that the lysine acetyltransferase Tip60 acetylates eEF1A1, whereas the histone deacetylase HDAC2 deacetylates eEF1A1. Promoting eEF1A1 deacetylation maintains the activation of Sox10 target genes and increases PNS and CNS remyelination efficiency. Taken together, these data identify a major mechanism of Sox10 regulation, which appears promising for future translational studies on PNS and CNS remyelination.


Asunto(s)
Factor 1 de Elongación Peptídica/metabolismo , Remielinización/genética , Activación Transcripcional/genética , Acetilación , Envejecimiento/metabolismo , Animales , Desdiferenciación Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/metabolismo , Lisina Acetiltransferasa 5/metabolismo , Ratones , Modelos Biológicos , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Sistema Nervioso Periférico/efectos de los fármacos , Sistema Nervioso Periférico/fisiología , Recuperación de la Función/efectos de los fármacos , Remielinización/efectos de los fármacos , Factores de Transcripción SOXE/metabolismo , Factor de Transcripción STAT3/metabolismo , Células de Schwann/efectos de los fármacos , Células de Schwann/metabolismo , Teofilina/farmacología , Transactivadores/metabolismo , Activación Transcripcional/efectos de los fármacos
19.
Cells ; 9(7)2020 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-32708794

RESUMEN

Multiple sclerosis (MS) is an autoimmune disorder characterised by demyelination of central nervous system neurons with subsequent damage, cell death and disability. While mechanisms exist in the CNS to repair this damage, they are disrupted in MS and currently there are no treatments to address this deficit. In recent years, increasing attention has been paid to the influence of the small, non-coding RNA molecules, microRNAs (miRNAs), in autoimmune disorders, including MS. In this review, we examine the role of miRNAs in remyelination in the different cell types that contribute to MS. We focus on key miRNAs that have a central role in mediating the repair process, along with several more that play either secondary or inhibitory roles in one or more aspects. Finally, we consider the current state of miRNAs as therapeutic targets in MS, acknowledging current challenges and potential strategies to overcome them in developing effective novel therapeutics to enhance repair mechanisms in MS.


Asunto(s)
MicroARNs/genética , Esclerosis Múltiple/genética , Esclerosis Múltiple/patología , Remielinización/genética , Animales , Sistema Nervioso Central/patología , Humanos , MicroARNs/inmunología , Modelos Biológicos , Terapia Molecular Dirigida , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/terapia
20.
J Exp Med ; 217(11)2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-32648893

RESUMEN

Wallerian degeneration (WD) is a process of autonomous distal degeneration of axons upon injury. Macrophages (MPs) of the peripheral nervous system (PNS) are the main cellular agent controlling this process. Some evidence suggests that resident PNS-MPs along with MPs of hematogenous origin may be involved, but whether these two subsets exert distinct functions is unknown. Combining MP-designed fluorescent reporter mice and coherent anti-Stokes Raman scattering (CARS) imaging of the sciatic nerve, we deciphered the spatiotemporal choreography of resident and recently recruited MPs after injury and unveiled distinct functions of these subsets, with recruited MPs being responsible for efficient myelin stripping and clearance and resident MPs being involved in axonal regrowth. This work provides clues to tackle selectively cellular processes involved in neurodegenerative diseases.


Asunto(s)
Macrófagos/inmunología , Degeneración Walleriana/diagnóstico por imagen , Degeneración Walleriana/inmunología , Animales , Axones/fisiología , Modelos Animales de Enfermedad , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Vaina de Mielina/fisiología , Microscopía Óptica no Lineal , Remielinización/genética , Nervio Ciático/diagnóstico por imagen , Nervio Ciático/inmunología , Nervio Ciático/lesiones , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA