Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 238
Filtrar
1.
Glia ; 72(8): 1469-1483, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38771121

RESUMEN

Myelination is the terminal step in a complex and precisely timed program that orchestrates the proliferation, migration and differentiation of oligodendroglial cells. It is thought that Sonic Hedgehog (Shh) acting on Smoothened (Smo) participates in regulating this process, but that these effects are highly context dependent. Here, we investigate oligodendroglial development and remyelination from three specific transgenic lines: NG2-CreERT2 (control), Smofl/fl/NG2-CreERT2 (loss of function), and SmoM2/NG2-CreERT2 (gain of function), as well as pharmacological manipulation that enhance or inhibit the Smo pathway (Smoothened Agonist (SAG) or cyclopamine treatment, respectively). To explore the effects of Shh/Smo on differentiation and myelination in vivo, we developed a highly quantifiable model by transplanting oligodendrocyte precursor cells (OPCs) in the retina. We find that myelination is greatly enhanced upon cyclopamine treatment and hypothesize that Shh/Smo could promote OPC proliferation to subsequently inhibit differentiation. Consistent with this hypothesis, we find that the genetic activation of Smo significantly increased numbers of OPCs and decreased oligodendrocyte differentiation when we examined the corpus callosum during development and after cuprizone demyelination and remyelination. However, upon loss of function with the conditional ablation of Smo, myelination in the same scenarios are unchanged. Taken together, our present findings suggest that the Shh pathway is sufficient to maintain OPCs in an undifferentiated state, but is not necessary for myelination and remyelination.


Asunto(s)
Diferenciación Celular , Proteínas Hedgehog , Ratones Transgénicos , Vaina de Mielina , Células Precursoras de Oligodendrocitos , Receptor Smoothened , Animales , Proteínas Hedgehog/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Células Precursoras de Oligodendrocitos/efectos de los fármacos , Receptor Smoothened/metabolismo , Receptor Smoothened/genética , Vaina de Mielina/metabolismo , Diferenciación Celular/fisiología , Diferenciación Celular/efectos de los fármacos , Alcaloides de Veratrum/farmacología , Ratones , Remielinización/fisiología , Remielinización/efectos de los fármacos , Oligodendroglía/metabolismo , Oligodendroglía/efectos de los fármacos , Oligodendroglía/fisiología , Ratones Endogámicos C57BL , Transducción de Señal/fisiología , Transducción de Señal/efectos de los fármacos
2.
Mol Cell Neurosci ; 129: 103937, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38796120

RESUMEN

Experimental models of multiple sclerosis (MS) have significantly contributed to our understanding of pathophysiology and the development of therapeutic interventions. Various in vivo animal models have successfully replicated key features of MS and associated pathophysiological processes, shedding light on the sequence of events leading to disease initiation, progression, and resolution. Nevertheless, these models often entail substantial costs and prolonged treatment periods. In contrast, in vitro models offer distinct advantages, including cost-effectiveness and precise control over experimental conditions, thereby facilitating more reproducible results. We have developed a novel in vitro model tailored to the study of oligodendroglial maturation and myelin deposition under demyelinating and remyelinating conditions, which encompasses all the cell types present in the central nervous system (CNS). Of note, our model enables the evaluation of microglial cell commitment through a protocol involving their depletion and subsequent repopulation. Given that the development and survival of microglia are critically reliant on colony-stimulating factor-1 receptor (CSF-1R) signaling, we have employed CSF-1R inhibition to effectively deplete microglia. This versatile model holds promise for the assessment of potential therapies aimed at promoting oligodendroglial differentiation to safeguard and repair myelin, hence mitigate neurodegenerative processes.


Asunto(s)
Microglía , Vaina de Mielina , Oligodendroglía , Remielinización , Microglía/metabolismo , Animales , Oligodendroglía/metabolismo , Vaina de Mielina/metabolismo , Ratones , Remielinización/fisiología , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/patología , Diferenciación Celular/fisiología , Células Cultivadas
3.
Bull Exp Biol Med ; 176(5): 666-671, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38727956

RESUMEN

This paper shows for the first time that co-transplantation of human olfactory ensheathing cells with neurotrophin-3 into spinal cord cysts is more effective for activation of remyelination than transplantation of cells with brain-derived neurotrophic factor and a combination of these two factors. The studied neurotrophic factors do not affect proliferation and migration of ensheathing cells in vitro. It can be concluded that the maximum improvement of motor function in rats receiving ensheathing cells with neurotrophin-3 is largely determined by activation of remyelination.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Neurotrofina 3 , Bulbo Olfatorio , Remielinización , Animales , Ratas , Neurotrofina 3/metabolismo , Humanos , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/farmacología , Remielinización/fisiología , Bulbo Olfatorio/citología , Proliferación Celular , Médula Espinal/metabolismo , Vaina de Mielina/metabolismo , Vaina de Mielina/fisiología , Células Cultivadas , Movimiento Celular , Quistes/patología , Femenino , Quistes del Sistema Nervioso Central/cirugía , Quistes del Sistema Nervioso Central/patología
4.
Glia ; 72(8): 1392-1401, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38572807

RESUMEN

Multiple sclerosis is an autoimmune disease of the central nervous system (CNS) characterized by demyelination, axonal damage and, for the majority of people, a decline in neurological function in the long-term. Remyelination could assist in the protection of axons and their functional recovery, but such therapies are not, as yet, available. The TAM (Tyro3, Axl, and MERTK) receptor ligand GAS6 potentiates myelination in vitro and promotes recovery in pre-clinical models of MS. However, it has remained unclear which TAM receptor is responsible for transducing this effect and whether post-translational modification of GAS6 is required. In this study, we show that the promotion of myelination requires post-translational modification of the GLA domain of GAS6 via vitamin K-dependent γ-carboxylation. We also confirmed that the intracerebroventricular provision of GAS6 for 2 weeks to demyelinated wild-type (WT) mice challenged with cuprizone increased the density of myelinated axons in the corpus callosum by over 2-fold compared with vehicle control. Conversely, the provision of GAS6 to Tyro3 KO mice did not significantly improve the density of myelinated axons. The improvement in remyelination following the provision of GAS6 to WT mice was also accompanied by an increased density of CC1+ve mature oligodendrocytes compared with vehicle control, whereas this improvement was not observed in the absence of Tyro3. This effect occurs independent of any influence on microglial activation. This work therefore establishes that the remyelinative activity of GAS6 is dependent on Tyro3 and includes potentiation of oligodendrocyte numbers.


Asunto(s)
Cuprizona , Enfermedades Desmielinizantes , Péptidos y Proteínas de Señalización Intercelular , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Tirosina Quinasas Receptoras , Remielinización , Animales , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/farmacología , Remielinización/fisiología , Remielinización/efectos de los fármacos , Proteínas Tirosina Quinasas Receptoras/metabolismo , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/patología , Cuprizona/toxicidad , Ratones , Modelos Animales de Enfermedad , Vaina de Mielina/metabolismo , Vaina de Mielina/efectos de los fármacos , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Masculino , Femenino
5.
Brain Stimul ; 17(3): 575-587, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38648972

RESUMEN

BACKGROUND: Current treatments for Multiple Sclerosis (MS) poorly address chronic innate neuroinflammation nor do they offer effective remyelination. The vagus nerve has a strong regulatory role in inflammation and Vagus Nerve Stimulation (VNS) has potential to affect both neuroinflammation and remyelination in MS. OBJECTIVE: This study investigated the effects of VNS on demyelination and innate neuroinflammation in a validated MS rodent model. METHODS: Lysolecithin (LPC) was injected in the corpus callosum (CC) of 46 Lewis rats, inducing a demyelinated lesion. 33/46 rats received continuously-cycled VNS (cVNS) or one-minute per day VNS (1minVNS) or sham VNS from 2 days before LPC-injection until perfusion at 3 days post-injection (dpi) (corresponding with a demyelinated lesion with peak inflammation). 13/46 rats received cVNS or sham from 2 days before LPC-injection until perfusion at 11 dpi (corresponding with a partial remyelinated lesion). Immunohistochemistry and proteomics analyses were performed to investigate the extend of demyelination and inflammation. RESULTS: Immunohistochemistry showed that cVNS significantly reduced microglial and astrocytic activation in the lesion and lesion border, and significantly reduced the Olig2+ cell count at 3 dpi. Furthermore, cVNS significantly improved remyelination with 57.4 % versus sham at 11 dpi. Proteomic gene set enrichment analyses showed increased activation of (glutamatergic) synapse pathways in cVNS versus sham, most pronounced at 3 dpi. CONCLUSION: cVNS improved remyelination of an LPC-induced lesion. Possible mechanisms might include modulation of microglia and astrocyte activity, increased (glutamatergic) synapses and enhanced oligodendrocyte clearance after initial injury.


Asunto(s)
Enfermedades Desmielinizantes , Lisofosfatidilcolinas , Ratas Endogámicas Lew , Remielinización , Estimulación del Nervio Vago , Animales , Ratas , Remielinización/fisiología , Remielinización/efectos de los fármacos , Lisofosfatidilcolinas/toxicidad , Enfermedades Desmielinizantes/terapia , Enfermedades Desmielinizantes/inducido químicamente , Estimulación del Nervio Vago/métodos , Masculino , Enfermedades Neuroinflamatorias/terapia , Enfermedades Neuroinflamatorias/inducido químicamente , Enfermedades Neuroinflamatorias/etiología , Modelos Animales de Enfermedad , Esclerosis Múltiple/terapia , Esclerosis Múltiple/inducido químicamente , Cuerpo Calloso
6.
Acta Neuropathol ; 147(1): 75, 2024 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-38656399

RESUMEN

In multiple sclerosis (MS), persisting disability can occur independent of relapse activity or development of new central nervous system (CNS) inflammatory lesions, termed chronic progression. This process occurs early and it is mostly driven by cells within the CNS. One promising strategy to control progression of MS is the inhibition of the enzyme Bruton's tyrosine kinase (BTK), which is centrally involved in the activation of both B cells and myeloid cells, such as macrophages and microglia. The benefit of BTK inhibition by evobrutinib was shown as we observed reduced pro-inflammatory activation of microglia when treating chronic experimental autoimmune encephalomyelitis (EAE) or following the adoptive transfer of activated T cells. Additionally, in a model of toxic demyelination, evobrutinib-mediated BTK inhibition promoted the clearance of myelin debris by microglia, leading to an accelerated remyelination. These findings highlight that BTK inhibition has the potential to counteract underlying chronic progression of MS.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa , Encefalomielitis Autoinmune Experimental , Microglía , Vaina de Mielina , Piperidinas , Pirimidinas , Animales , Femenino , Ratones , Agammaglobulinemia Tirosina Quinasa/antagonistas & inhibidores , Agammaglobulinemia Tirosina Quinasa/metabolismo , Compuestos de Bifenilo/farmacología , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/patología , Ratones Endogámicos C57BL , Microglía/patología , Microglía/efectos de los fármacos , Microglía/metabolismo , Vaina de Mielina/patología , Vaina de Mielina/metabolismo , Piperidinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Remielinización/fisiología , Remielinización/efectos de los fármacos
7.
J Neuroophthalmol ; 44(2): 143-156, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38654413

RESUMEN

INTRODUCTION: Amelioration of disability in multiple sclerosis requires the development of complementary therapies that target neurodegeneration and promote repair. Remyelination is a promising neuroprotective strategy that may protect axons from damage and subsequent neurodegeneration. METHODS: A review of key literature plus additional targeted search of PubMed and Google Scholar was conducted. RESULTS: There has been a rapid expansion of clinical trials studying putative remyelinating candidates, but further growth of the field is limited by the lack of consensus on key aspects of trial design. We have not yet defined the ideal study population, duration of therapy, or the appropriate outcome measures to detect remyelination in humans. The varied natural history of multiple sclerosis, coupled with the short time frame of phase II clinical trials, requires that we develop and validate biomarkers of remyelination that can serve as surrogate endpoints in clinical trials. CONCLUSIONS: We propose that the visual system may be the most well-suited and validated model for the study potential remyelinating agents. In this review, we discuss the pathophysiology of demyelination and summarize the current clinical trial landscape of remyelinating agents. We present some of the challenges in the study of remyelinating agents and discuss current potential biomarkers of remyelination and repair, emphasizing both established and emerging visual outcome measures.


Asunto(s)
Esclerosis Múltiple , Remielinización , Humanos , Esclerosis Múltiple/fisiopatología , Esclerosis Múltiple/tratamiento farmacológico , Remielinización/fisiología , Remielinización/efectos de los fármacos , Vaina de Mielina
8.
Nat Commun ; 15(1): 1870, 2024 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-38467607

RESUMEN

Myelin regeneration (remyelination) is essential to prevent neurodegeneration in demyelinating diseases such as Multiple Sclerosis, however, its efficiency declines with age. Regulatory T cells (Treg) recently emerged as critical players in tissue regeneration, including remyelination. However, the effect of ageing on Treg-mediated regenerative processes is poorly understood. Here, we show that expansion of aged Treg does not rescue age-associated remyelination impairment due to an intrinsically diminished capacity of aged Treg to promote oligodendrocyte differentiation and myelination in male and female mice. This decline in regenerative Treg functions can be rescued by a young environment. We identified Melanoma Cell Adhesion Molecule 1 (MCAM1) and Integrin alpha 2 (ITGA2) as candidates of Treg-mediated oligodendrocyte differentiation that decrease with age. Our findings demonstrate that ageing limits the neuroregenerative capacity of Treg, likely limiting their remyelinating therapeutic potential in aged patients, and describe two mechanisms implicated in Treg-driven remyelination that may be targetable to overcome this limitation.


Asunto(s)
Remielinización , Humanos , Masculino , Femenino , Ratones , Animales , Anciano , Remielinización/fisiología , Linfocitos T Reguladores/metabolismo , Oligodendroglía/fisiología , Diferenciación Celular/fisiología , Vaina de Mielina/metabolismo , Envejecimiento , Sistema Nervioso Central
9.
Neurorehabil Neural Repair ; 38(5): 350-363, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38491852

RESUMEN

BACKGROUND: Yi-Qi-Tong-Luo Granules (YQTLs) is a natural compound of Traditional Chinese Medicine authorized by China Food and Drug Administration (CFDA). These granules are employed in the convalescent stage of cerebral infarction and render notable clinical efficacy. This study aims to uncover the underlying mechanisms of YQTLs on remyelination after cerebral ischemia injury. MATERIALS AND METHODS: We established cerebral ischemia model in rats using microsphere-induced multiple cerebral infarction (MCI). We evaluated the pharmacological effects of YQTLs on MCI rats, through Morri's water maze test, open field test, hematoxylin and eosin staining, and glycine silver immersion. We employed liquid chromatography mass spectrometry metabolomics to identify differential metabolites. Enzyme-linked immunosorbent assay was utilized to measure the release of neurotrophins, while immunofluorescence staining was used to assess oligodendrocyte precursor cells differences and myelin regeneration. We used Western blotting to validate the protein expression of remyelination-associated signaling pathways. RESULTS: YQTLs significantly improves cognitive function following cerebral ischemia injury. Pathological tissue staining revealed that YQTLs administration inhibits neuronal denaturation and neurofibrillary tangles. We identified 141 differential metabolites among the sham, MCI, and YQTLs-treated MCI groups. Among these metabolites, neurotransmitters were identified, and notably, gamma-aminobutyric acid (GABA) showed marked improvement in the YQTLs group. The induction of neurotrophins, such as brain-derived neurotrophic factor (BDNF) and PDGFAA, upregulation of olig2 and MBP expression, and promotion of remyelination were evident in YQTLs-treated MCI groups. Gamma-aminobutyric acid B receptors (GABABR), pERK/extracellular regulated MAP kinase, pAKT/protein kinase B, and pCREB/cAMP response element-binding were upregulated following YQTLs treatment. CONCLUSION: YQTLs enhance the binding of GABA to GABABR, thereby activating the pCREB/BDNF signaling pathway, which in turn increases the expression of downstream myelin-associated proteins and promotes remyelination and cognitive function.


Asunto(s)
Isquemia Encefálica , Factor Neurotrófico Derivado del Encéfalo , Metabolómica , Ratas Sprague-Dawley , Remielinización , Transducción de Señal , Animales , Remielinización/efectos de los fármacos , Remielinización/fisiología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/efectos de los fármacos , Ratas , Masculino , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/efectos de los fármacos
10.
Artículo en Inglés | MEDLINE | ID: mdl-38316552

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, while this is largely true of the neuronal elements of the adult mammalian CNS, save for discrete populations of granule 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 work, we will (1) review the biology of remyelination, including the cells and signals involved; (2) describe when remyelination occurs and when and why it fails, including the consequences of its failure; and (3) 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)
Enfermedades Desmielinizantes , Remielinización , Animales , Adulto , Niño , Humanos , Remielinización/fisiología , Regeneración Nerviosa/fisiología , Vaina de Mielina/fisiología , Sistema Nervioso Central , Mamíferos
11.
Brain Pathol ; 34(2): e13218, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-37927164

RESUMEN

Multiple sclerosis is a chronic autoimmune disease of the central nervous system characterized by myelin loss, axonal damage, and glial scar formation. Still, the underlying processes remain unclear, as numerous pathways and factors have been found to be involved in the development and progression of the disease. Therefore, it is of great importance to find suitable animal models as well as reliable methods for their precise and reproducible analysis. Here, we describe the impact of demyelination on clinically relevant gray matter regions of the hippocampus and cerebral cortex, using the previously established cuprizone model for aged mice. We could show that bioinformatic image analysis methods are not only suitable for quantification of cell populations, but also for the assessment of de- and remyelination processes, as numerous objective parameters can be considered for reproducible measurements. After cuprizone-induced demyelination, subsequent remyelination proceeded slowly and remained incomplete in all gray matter areas studied. There were regional differences in the number of mature oligodendrocytes during remyelination suggesting region-specific differences in the factors accounting for remyelination failure, as, even in the presence of oligodendrocytes, remyelination in the cortex was found to be impaired. Upon cuprizone administration, synaptic density and dendritic volume in the gray matter of aged mice decreased. The intensity of synaptophysin staining gradually restored during the subsequent remyelination phase, however the expression of MAP2 did not fully recover. Microgliosis persisted in the gray matter of aged animals throughout the remyelination period, whereas extensive astrogliosis was of short duration as compared to white matter structures. In conclusion, we demonstrate that the application of the cuprizone model in aged mice mimics the impaired regeneration ability seen in human pathogenesis more accurately than commonly used protocols with young mice and therefore provides an urgently needed animal model for the investigation of remyelination failure and remyelination-enhancing therapies.


Asunto(s)
Enfermedades Desmielinizantes , Remielinización , Humanos , Ratones , Animales , Anciano , Cuprizona/toxicidad , Sustancia Gris/patología , Enfermedades Desmielinizantes/patología , Remielinización/fisiología , Corteza Cerebral/patología , Oligodendroglía/patología , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Vaina de Mielina/patología
12.
Brain ; 147(5): 1871-1886, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38128553

RESUMEN

Multiple sclerosis is a chronic inflammatory disease in which disability results from the disruption of myelin and axons. During the initial stages of the disease, injured myelin is replaced by mature myelinating oligodendrocytes that differentiate from oligodendrocyte precursor cells. However, myelin repair fails in secondary and chronic progressive stages of the disease and with ageing, as the environment becomes progressively more hostile. This may be attributable to inhibitory molecules in the multiple sclerosis environment including activation of the p38MAPK family of kinases. We explored oligodendrocyte precursor cell differentiation and myelin repair using animals with conditional ablation of p38MAPKγ from oligodendrocyte precursors. We found that p38γMAPK ablation accelerated oligodendrocyte precursor cell differentiation and myelination. This resulted in an increase in both the total number of oligodendrocytes and the migration of progenitors ex vivo and faster remyelination in the cuprizone model of demyelination/remyelination. Consistent with its role as an inhibitor of myelination, p38γMAPK was significantly downregulated as oligodendrocyte precursor cells matured into oligodendrocytes. Notably, p38γMAPK was enriched in multiple sclerosis lesions from patients. Oligodendrocyte progenitors expressed high levels of p38γMAPK in areas of failed remyelination but did not express detectable levels of p38γMAPK in areas where remyelination was apparent. Our data suggest that p38γ could be targeted to improve myelin repair in multiple sclerosis.


Asunto(s)
Esclerosis Múltiple , Vaina de Mielina , Oligodendroglía , Remielinización , Animales , Remielinización/fisiología , Esclerosis Múltiple/patología , Esclerosis Múltiple/metabolismo , Vaina de Mielina/metabolismo , Vaina de Mielina/patología , Ratones , Oligodendroglía/metabolismo , Oligodendroglía/patología , Humanos , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Proteína Quinasa 12 Activada por Mitógenos/genética , Diferenciación Celular/fisiología , Cuprizona/toxicidad , Ratones Endogámicos C57BL , Masculino , Femenino , Enfermedades Desmielinizantes/patología , Enfermedades Desmielinizantes/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Células Precursoras de Oligodendrocitos/patología , Ratones Transgénicos
13.
J Neuroimmunol ; 384: 578219, 2023 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-37813042

RESUMEN

Small ubiquitin like modifiers (SUMO) are reversible posttranslational modifiers of intracellular proteins. In the CNS, expression of myelin genes is regulated by state of SUMOylation of their respective transcription factors. In the immune system, deSUMOylation activates innate immune responses and promotes anti-viral immunity. However, the role played by SUMO in an adaptive immune response and in the development of T cell mediated autoimmune disease has not been previously described. TAK981 is a synthetic small molecule which by forming adducts with SUMO proteins prevents SUMOylation. We examined the expression of myelin genes and their transcription factors following culture with TAK981 in Oligodendrocyte Precursor Cells (OPC). We found that myelin basic protein (MBP), a key myelin protein, is upregulated in OPC in the presence of TAK981. We also found increased expression of transcription factors Sox10 and Myrf, which engage in the expression of MBP. In the Cuprizone model of demyelination/remyelination, animals which were treated with TAK981 showed increased remyelination in areas of demyelination and an increase in the number of maturing oligodendrocytes compared to vehicle treated controls. In in vitro cultures of lymphocytes, TAK981 reduced the expression of TH17 in T cells in mice immunized with MOGp35-55. Following in vivo treatment with TAK981, there was a significant reduction in the clinical and pathological severity in mice immunized to develop experimental allergic encephalitis (EAE). The dual effects of deSUMOylation on remyelination and in regulating an autoimmune adaptive response offers a novel approach to the management of human inflammatory demyelinating diseases such as multiple sclerosis.


Asunto(s)
Enfermedades del Sistema Nervioso Central , Enfermedades Desmielinizantes , Remielinización , Ratones , Humanos , Animales , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/tratamiento farmacológico , Enfermedades Desmielinizantes/metabolismo , Remielinización/fisiología , Sumoilación , Interleucina-17 , Diferenciación Celular , Vaina de Mielina/patología , Oligodendroglía/metabolismo , Cuprizona/toxicidad , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Enfermedades del Sistema Nervioso Central/metabolismo , Factores de Transcripción/metabolismo , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad
14.
ASN Neuro ; 15: 17590914231167281, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37654230

RESUMEN

SUMMARY STATEMENT: Bone marrow cell transplant has proven to be an effective therapeutic approach to treat peripheral nervous system injuries as it not only promoted regeneration and remyelination of the injured nerve but also had a potent effect on neuropathic pain.


Asunto(s)
Axones , Remielinización , Sistema Nervioso Periférico , Regeneración Nerviosa/fisiología , Remielinización/fisiología , Células de la Médula Ósea
16.
Mol Neurobiol ; 60(9): 5493-5504, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37322287

RESUMEN

Ischemic stroke is one of the main reasons of disability and death. Stroke-induced functional deficits are mainly due to the secondary degeneration of the white matter characterized by axonal demyelination and injury of axon-glial integrity. Enhancement of the axonal regeneration and remyelination could promote the neural functional recovery. However, cerebral ischemia-induced activation of RhoA/Rho kinase (ROCK) pathway plays a crucial and harmful role in the process of axonal recovery and regeneration. Inhibition of this pathway could promote the axonal regeneration and remyelination. In addition, hydrogen sulfide (H2S) has the significant neuroprotective role during the recovery of ischemic stroke via inhibiting the inflammatory response and oxidative stress, regulating astrocyte function, promoting the differentiation of endogenous oligodendrocyte precursor cells (OPCs) to mature oligodendrocyte. Among all of these effects, promoting the formation of mature oligodendrocyte is a crucial part of axonal regeneration and remyelination. Furthermore, numerous studies have uncovered the crosstalk between astrocytes and oligodendrocyte, microglial cells and oligodendrocyte in the axonal remyelination following ischemic stroke. The purpose of this review was to discuss the relationship among H2S, RhoA/ROCK pathway, astrocytes, and microglial cells in the axonal remyelination following ischemic stroke to reveal new strategies for preventing and treating this devastating disease.


Asunto(s)
Accidente Cerebrovascular Isquémico , Remielinización , Accidente Cerebrovascular , Humanos , Remielinización/fisiología , Accidente Cerebrovascular Isquémico/metabolismo , Quinasas Asociadas a rho/metabolismo , Neuroglía , Oligodendroglía/metabolismo , Axones , Accidente Cerebrovascular/metabolismo , Diferenciación Celular
17.
Prog Neurobiol ; 226: 102459, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37127087

RESUMEN

Oligodendrocytes are responsible for myelinating central nervous system (CNS) axons and rapid electrical transmission through saltatory conduction of action potentials. Myelination and myelin repair rely partially on oligodendrogenesis, which comprises oligodendrocyte precursor cell (OPC) migration, maturation, and differentiation into oligodendrocytes (OL). In multiple sclerosis (MS), demyelination occurs due to an inflammatory cascade with auto-reactive T-cells. When oligodendrogenesis fails, remyelination becomes aberrant and conduction impairments are no longer restored. Although current disease modifying therapies have achieved results in modulating the faulty immune response, disease progression continues because of chronic inflammation, neurodegeneration, and failure of remyelination. Therapies have been tried to promote remyelination. Modulation of neuronal activity seems to be a very promising strategy in preclinical studies. Additionally, studies in people with MS (pwMS) have shown symptom improvement following non-invasive brain stimulation. (NIBS) techniques. The aforementioned mechanisms are yet unknown and probably involve both the activation of neurons and glial cells. Noting neuronal activity contributes to myelin plasticity and that NIBS modulates neuronal activity; we argue that NIBS is a promising research horizon for demyelinating diseases. We review the hypothesized pathways through which NIBS may affect both neuronal activity in the CNS and how the resulting activity can affect oligodendrogenesis and myelination.


Asunto(s)
Esclerosis Múltiple , Remielinización , Humanos , Remielinización/fisiología , Vaina de Mielina/fisiología , Oligodendroglía/fisiología , Esclerosis Múltiple/terapia , Esclerosis Múltiple/metabolismo , Diferenciación Celular/fisiología
18.
Trends Neurosci ; 46(7): 581-596, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37183154

RESUMEN

Adult oligodendrocyte precursor cells (aOPCs), transformed from fetal OPCs, are idiosyncratic neuroglia of the central nervous system (CNS) that are distinct in many ways from other glial cells. OPCs have been classically studied in the context of their remyelinating capacity. Recent studies, however, revealed that aOPCs not only contribute to post-lesional remyelination but also play diverse crucial roles in multiple neurological diseases. In this review we briefly present the physiology of aOPCs and summarize current knowledge of the beneficial and detrimental roles of aOPCs in different CNS diseases. We discuss unique features of aOPC death, reactivity, and changes during senescence, as well as aOPC interactions with other glial cells and pathological remodeling during disease. Finally, we outline future perspectives for the study of aOPCs in brain pathologies which may instigate the development of aOPC-targeting therapeutic strategies.


Asunto(s)
Células Precursoras de Oligodendrocitos , Remielinización , Células Precursoras de Oligodendrocitos/fisiología , Sistema Nervioso Central , Neuroglía , Remielinización/fisiología , Oligodendroglía/fisiología , Diferenciación Celular/fisiología , Vaina de Mielina/fisiología
19.
Neuropeptides ; 100: 102348, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37236132

RESUMEN

Patients with a spinal cord injury (SCI) usually suffer lifelong disability as a result. Considering this, SCI treatment and pathology study are urgently needed. Metformin, a widely used hypoglycemic drug, has been indicated for its important role in central nervous system diseases. This study aimed to investigate the potential effect of metformin on remyelination after SCI. In the present study, we established a cervical contusion SCI model and metformin treatment was applied after SCI. Biomechanical parameters and behavioral assessment were used to evaluate the severity of injury and the improvement of functional recovery after SCI, respectively. The immunofluorescence and western blot were performed at the terminal time point. Our results showed that treating with metformin after SCI improved functional recovery by reducing the white matter loss and promoting Schwann cell remyelination, and the Nrg1/ErbB signaling pathway may be involved in promoting remyelination mediated by oligodendrocytes and Schwann cells. In addition, the area of spared tissues was significantly increased in the metformin group. However, metformin had no significant effects on the glial scar and inflammation after SCI. In summary, these findings indicated that the role of metformin in Schwann cell remyelination after SCI was probably related to the regulation of the Nrg1/ErbB pathway. It is, therefore, possible to suggest that metformin may be a potential therapy for SCI.


Asunto(s)
Metformina , Remielinización , Traumatismos de la Médula Espinal , Humanos , Remielinización/fisiología , Metformina/uso terapéutico , Metformina/metabolismo , Metformina/farmacología , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/patología , Células de Schwann/metabolismo , Células de Schwann/patología , Oligodendroglía/patología , Médula Espinal/metabolismo , Recuperación de la Función/fisiología
20.
Cells ; 12(9)2023 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-37174731

RESUMEN

Spinal cord injury (SCI) is characterized by a cascade of events that lead to sensory and motor disabilities. To date, this condition is irreversible, and no cure exists. To improve myelin repair and limit secondary degeneration, we developed a multitherapy based on nanomedicines (NMeds) loaded with the promyelinating agent triiodothyronine (T3), used in combination with systemic ibuprofen and mouse nerve growth factor (mNGF). Poly-L-lactic-co-glycolic acid (PLGA) NMeds were optimized and loaded with T3 to promote sustained release. In vitro experiments confirmed the efficacy of T3-NMeds to differentiate oligodendrocyte precursor cells. In vivo rat experiments were performed in contusion SCI to explore the NMed biodistribution and efficacy of combo drugs at short- and long-term post-lesion. A strong anti-inflammatory effect was observed in the short term with a reduction of type M1 microglia and glutamate levels, but with a subsequent increase of TREM2. In the long term, an improvement of myelination in NG2-IR, an increase in MBP content, and a reduction of the demyelination area were observed. These data demonstrated that NMeds can successfully be used to obtain more controlled local drug delivery and that this multiple treatment could be effective in improving the outcome of SCIs.


Asunto(s)
Remielinización , Traumatismos de la Médula Espinal , Ratas , Ratones , Animales , Remielinización/fisiología , Distribución Tisular , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/patología , Vaina de Mielina/patología , Inflamación/tratamiento farmacológico , Inflamación/patología , Glicoproteínas de Membrana/farmacología , Receptores Inmunológicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...