Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 252
Filtrar
1.
Acta Neuropathol Commun ; 12(1): 154, 2024 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-39300502

RESUMEN

Aging is the greatest known risk factor for most neurodegenerative diseases. Myelin degeneration is an early pathological indicator of these diseases and a normal part of aging; albeit, to a lesser extent. Despite this, little is known about the contribution of age-related myelin degeneration on neurodegenerative disease. Microglia participate in modulating white matter events from demyelination to remyelination, including regulation of (de)myelination by the microglial innate immune receptor triggering receptor expressed on myeloid cells 2 (TREM2). Here, we demonstrate Trem2-deficiency aggravates and accelerates age-related myelin degeneration in the striatum. We show TREM2 is necessary for remyelination by recruiting reparative glia and mediating signaling that promotes OPC differentiation/maturation. In response to demyelination, TREM2 is required for phagocytosis of large volumes of myelin debris. In addition to lysosomal regulation, we show TREM2 can modify the ER stress response, even prior to overt myelin debris, that prevents lipid accumulation and microglial dysfunction. These data support a role for Trem2-dependent interactions in age-related myelin degeneration and suggest a basis for how early dysfunctional microglia could contribute to disease pathology through insufficent repair, defective phagocytosis, and the ER stress response.


Asunto(s)
Envejecimiento , Glicoproteínas de Membrana , Microglía , Vaina de Mielina , Receptores Inmunológicos , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/deficiencia , Animales , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/deficiencia , Vaina de Mielina/patología , Vaina de Mielina/metabolismo , Envejecimiento/patología , Envejecimiento/metabolismo , Ratones , Microglía/metabolismo , Microglía/patología , Enfermedades Desmielinizantes/patología , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/genética , Ratones Endogámicos C57BL , Fagocitosis/genética , Ratones Noqueados , Estrés del Retículo Endoplásmico/fisiología , Remielinización/fisiología
2.
J Physiol Investig ; 67(4): 161-173, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-39175192

RESUMEN

ABSTRACT: Multiple sclerosis (MS) is a complicated, inflammatory disease that causes demyelination of the central nervous system (CNS), resulting in a variety of neurological abnormalities. Over the past several decades, different animal models have been used to replicate the clinical symptoms and neuropathology of MS. The experimental model of experimental autoimmune/allergic encephalomyelitis (EAE) and viral and toxin-induced model was widely used to investigate the clinical implications of MS. Discoidin domain receptor 1 (DDR1) signaling in oligodendrocytes (OL) brings a new dimension to our understanding of MS pathophysiology. DDR1 is effectively involved in the OL during neurodevelopment and remyelination. It has been linked to many cellular processes, including migration, invasion, proliferation, differentiation, and adhesion. However, the exact functional involvement of DDR1 in developing OL and myelinogenesis in the CNS remains undefined. In this review, we critically evaluate the current literature on DDR1 signaling in OL and its proliferation, migration, differentiation, and myelination mechanism in OL in association with the progression of MS. It increases our knowledge of DDR1 in OL as a novel target molecule for oligodendrocyte-associated diseases in the CNS, including MS.


Asunto(s)
Sistema Nervioso Central , Receptor con Dominio Discoidina 1 , Vaina de Mielina , Oligodendroglía , Remielinización , Transducción de Señal , Animales , Humanos , Diferenciación Celular , Sistema Nervioso Central/metabolismo , Receptor con Dominio Discoidina 1/metabolismo , Receptor con Dominio Discoidina 1/genética , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo , Remielinización/fisiología
3.
CNS Neurosci Ther ; 30(8): e14903, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39139089

RESUMEN

INTRODUCTION: Excessive neuroinflammation, apoptosis, glial scar, and demyelination triggered by spinal cord injury (SCI) are major obstacles to SCI repair. Fucoidan, a natural marine plant extract, possesses broad-spectrum anti-inflammatory and immunomodulatory effects and is regarded as a potential therapeutic for various diseases, including neurological disorders. However, its role in SCI has not been investigated. METHODS: In this study, we established an SCI model in mice and intervened in injury repair by daily intraperitoneal injections of different doses of fucoidan (10 and 20 mg/kg). Concurrently, primary oligodendrocyte precursor cells (OPCs) were treated in vitro to validate the differentiation-promoting effect of fucoidan on OPCs. Basso Mouse Scale (BMS), Louisville Swim Scale (LSS), and Rotarod test were carried out to measure the functional recovery. Immunofluorescence staining, and transmission electron microscopy (TEM) were performed to assess the neuroinflammation, apoptosis, glial scar, and remyelination. Western blot analysis was conducted to clarify the underlying mechanism of remyelination. RESULTS: Our results indicate that in the SCI model, fucoidan exhibits significant anti-inflammatory effects and promotes the transformation of pro-inflammatory M1-type microglia/macrophages into anti-inflammatory M2-type ones. Fucoidan enhances the survival of neurons and axons in the injury area and improves remyelination. Additionally, fucoidan promotes OPCs differentiation into mature oligodendrocytes by activating the PI3K/AKT/mTOR pathway. CONCLUSION: Fucoidan improves SCI repair by modulating the microenvironment and promoting remyelination.


Asunto(s)
Ratones Endogámicos C57BL , Polisacáridos , Recuperación de la Función , Remielinización , Traumatismos de la Médula Espinal , Animales , Polisacáridos/farmacología , Ratones , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/patología , Remielinización/efectos de los fármacos , Remielinización/fisiología , Recuperación de la Función/efectos de los fármacos , Células Precursoras de Oligodendrocitos/efectos de los fármacos , Femenino , Microambiente Celular/efectos de los fármacos
4.
Elife ; 122024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39163103

RESUMEN

Revealing unknown cues that regulate oligodendrocyte progenitor cell (OPC) function in remyelination is important to optimise the development of regenerative therapies for multiple sclerosis (MS). Platelets are present in chronic non-remyelinated lesions of MS and an increase in circulating platelets has been described in experimental autoimmune encephalomyelitis (EAE) mice, an animal model for MS. However, the contribution of platelets to remyelination remains unexplored. Here we show platelet aggregation in proximity to OPCs in areas of experimental demyelination. Partial depletion of circulating platelets impaired OPC differentiation and remyelination, without altering blood-brain barrier stability and neuroinflammation. Transient exposure to platelets enhanced OPC differentiation in vitro, whereas sustained exposure suppressed this effect. In a mouse model of thrombocytosis (Calr+/-), there was a sustained increase in platelet aggregation together with a reduction of newly-generated oligodendrocytes following toxin-induced demyelination. These findings reveal a complex bimodal contribution of platelet to remyelination and provide insights into remyelination failure in MS.


Asunto(s)
Plaquetas , Diferenciación Celular , Células Precursoras de Oligodendrocitos , Remielinización , Animales , Células Precursoras de Oligodendrocitos/fisiología , Remielinización/fisiología , Ratones , Plaquetas/fisiología , Encefalomielitis Autoinmune Experimental/patología , Ratones Endogámicos C57BL , Esclerosis Múltiple/patología , Modelos Animales de Enfermedad , Oligodendroglía/fisiología , Femenino
5.
Adv Neurobiol ; 37: 445-456, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39207707

RESUMEN

Multiple sclerosis (MS) is a devastating autoimmune disease that leads to profound disability. This disability arises from the stochastic, regional loss of myelin-the insulating sheath surrounding neurons-in the central nervous system (CNS). The demyelinated regions are dominated by the brain's resident macrophages: microglia. Microglia perform a variety of functions in MS and are thought to initiate and perpetuate demyelination through their interactions with peripheral immune cells that traffic into the brain. However, microglia are also likely essential for recruiting and promoting the differentiation of cells that can restore lost myelin in a process known as remyelination. Given these seemingly opposing functions, an overarching beneficial or detrimental role is yet to be ascribed to these immune cells. In this chapter, we will discuss microglia dynamics throughout the MS disease course and probe the apparent dichotomy of microglia as the drivers of both demyelination and remyelination.


Asunto(s)
Microglía , Esclerosis Múltiple , Vaina de Mielina , Microglía/metabolismo , Microglía/patología , Humanos , Esclerosis Múltiple/patología , Esclerosis Múltiple/inmunología , Vaina de Mielina/patología , Vaina de Mielina/metabolismo , Remielinización/fisiología , Animales , Encéfalo/patología , Encéfalo/inmunología , Enfermedades Desmielinizantes/patología , Enfermedades Desmielinizantes/inmunología , Enfermedades Desmielinizantes/metabolismo
7.
Neurology ; 103(6): e209604, 2024 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-39213476

RESUMEN

BACKGROUND AND OBJECTIVES: Myelin and iron play essential roles in remyelination processes of multiple sclerosis (MS) lesions. χ-separation, a novel biophysical model applied to multiecho T2*-data and T2-data, estimates the contribution of myelin and iron to the obtained susceptibility signal. We used this method to investigate myelin and iron levels in lesion and nonlesion brain areas in patients with MS and healthy individuals. METHODS: This prospective MS cohort study included patients with MS fulfilling the McDonald Criteria 2017 and healthy individuals, aged 18 years or older, with no other neurologic comorbidities. Participants underwent MRI at baseline and after 2 years, including multiecho GRE-(T2*) and FAST-(T2) sequences. Using χ-separation, we generated myelin-sensitive and iron-sensitive susceptibility maps. White matter lesions (WMLs), cortical lesions (CLs), surrounding normal-appearing white matter (NAWM), and normal-appearing gray matter were segmented on fluid-attenuated inversion recovery and magnetization-prepared 2 rapid gradient echo images, respectively. Cross-sectional group comparisons used Wilcoxon rank-sum tests, longitudinal analyses applied Wilcoxon signed-rank tests. Associations with clinical outcomes (disease phenotype, age, sex, disease duration, disability measured by Expanded Disability Status Scale [EDSS], neurofilament light chain levels, and T2-lesion number and volume) were assessed using linear regression models. RESULTS: Of 168 patients with MS (median [interquartile range (IQR)] age 47.0 [21.7] years; 101 women; 6,898 WMLs, 775 CLs) and 103 healthy individuals (age 33.0 [10.5] years, 57 women), 108 and 62 were followed for a median of 2 years, respectively (IQR 0.1; 5,030 WMLs, 485 CLs). At baseline, WMLs had lower myelin (median 0.025 [IQR 0.015] parts per million [ppm]) and iron (0.017 [0.015] ppm) than the corresponding NAWM (myelin 0.030 [0.012]; iron 0.019 [0.011] ppm; both p < 0.001). After 2 years, both myelin (0.027 [0.014] ppm) and iron had increased (0.018 [0.015] ppm; both p < 0.001). Younger age (p < 0.001, b = -5.111 × 10-5), lower disability (p = 0.04, b = -2.352 × 10-5), and relapsing-remitting phenotype (RRMS, 0.003 [0.01] vs primary progressive 0.002 [IQR 0.01], p < 0.001; vs secondary progressive 0.0004 [IQR 0.01], p < 0.001) at baseline were associated with remyelination. Increment of myelin correlated with clinical improvement measured by EDSS (p = 0.015, b = -6.686 × 10-4). DISCUSSION: χ-separation, a novel mathematical model applied to multiecho T2*-images and T2-images shows that young RRMS patients with low disability exhibit higher remyelination capacity, which correlated with clinical disability over a 2-year follow-up.


Asunto(s)
Imagen por Resonancia Magnética , Esclerosis Múltiple , Remielinización , Sustancia Blanca , Humanos , Femenino , Masculino , Adulto , Sustancia Blanca/diagnóstico por imagen , Sustancia Blanca/patología , Remielinización/fisiología , Persona de Mediana Edad , Esclerosis Múltiple/diagnóstico por imagen , Esclerosis Múltiple/patología , Estudios Prospectivos , Vaina de Mielina/patología , Hierro/metabolismo , Estudios Transversales , Corteza Cerebral/diagnóstico por imagen , Corteza Cerebral/patología , Estudios de Cohortes
8.
Mol Neurodegener ; 19(1): 53, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-38997755

RESUMEN

BACKGROUND: Multiple sclerosis (MS) therapeutic goals have traditionally been dichotomized into two distinct avenues: immune-modulatory-centric interventions and pro-regenerative strategies. Oligodendrocyte progenitor cells (OPCs) were regarded for many years solely in concern to their potential to generate oligodendrocytes and myelin in the central nervous system (CNS). However, accumulating data elucidate the multifaceted roles of OPCs, including their immunomodulatory functions, positioning them as cardinal constituents of the CNS's immune landscape. MAIN BODY: In this review, we will discuss how the two therapeutic approaches converge. We present a model by which (1) an inflammation is required for the appropriate pro-myelinating immune function of OPCs in the chronically inflamed CNS, and (2) the immune function of OPCs is crucial for their ability to differentiate and promote remyelination. This model highlights the reciprocal interactions between OPCs' pro-myelinating and immune-modulating functions. Additionally, we review the specific effects of anti- and pro-inflammatory interventions on OPCs, suggesting that immunosuppression adversely affects OPCs' differentiation and immune functions. CONCLUSION: We suggest a multi-systemic therapeutic approach, which necessitates not a unidimensional focus but a harmonious balance between OPCs' pro-myelinating and immune-modulatory functions.


Asunto(s)
Inflamación , Esclerosis Múltiple , Células Precursoras de Oligodendrocitos , Remielinización , Humanos , Remielinización/fisiología , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/terapia , Esclerosis Múltiple/patología , Animales , Inflamación/inmunología , Diferenciación Celular/fisiología , Vaina de Mielina , Oligodendroglía
9.
Mech Ageing Dev ; 220: 111959, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38950628

RESUMEN

Oligodendrocyte precursor cells (OPCs) comprise 5-8 % of the adult glial cell population and stand out as the most proliferative cell type in the central nervous system (CNS). OPCs are responsible for generating oligodendrocytes (OLs), the myelinating cells of the CNS. However, OPC functions decline as we age, resulting in impaired differentiation and inadequate remyelination. This review explores the cellular and molecular changes associated with OPC aging, and their impact on OPC differentiation and functionality. Furthermore, it examines the impact of OPC aging within the context of multiple sclerosis and Alzheimer's disease, both neurodegenerative conditions wherein aged OPCs exacerbate disease progression by impeding remyelination. Moreover, various pharmacological interventions targeting pathways related to senescence and differentiation are discussed as potential strategies to rejuvenate aged OPCs. Enhancing our understanding of OPC aging mechanisms holds promise for developing new therapies to improve remyelination and repair in age-related neurodegenerative disorders.


Asunto(s)
Encéfalo , Diferenciación Celular , Senescencia Celular , Células Precursoras de Oligodendrocitos , Humanos , Células Precursoras de Oligodendrocitos/metabolismo , Células Precursoras de Oligodendrocitos/fisiología , Senescencia Celular/fisiología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Diferenciación Celular/fisiología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Esclerosis Múltiple/patología , Esclerosis Múltiple/metabolismo , Envejecimiento/fisiología , Envejecimiento/metabolismo , Envejecimiento/patología , Oligodendroglía/metabolismo , Remielinización/fisiología
10.
Neuroscience ; 555: 41-51, 2024 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-39033991

RESUMEN

The research aims to study the therapeutic impact of HEK293-XPack-Olig2 cell-derived exosomes on remyelination of the corpus callosum in a cuprizone-induced demyelinating disease model. A lentiviral vector expressing Olig2 was constructed using XPack technology. The highly abundant Olig2 exosomes (ExoOs) were isolated by centrifugation for subsequent experiments. Western blot, nanoparticle tracking analysis (NTA), and electron microscopy showed no significant difference in particle size and morphology between Exos and ExoOs, and a high level of Olig2 expression could be detected in ExoOs, indicating that exosome modification by XPack technology was successful. The Black Gold/Fluromyelin staining analysis showed that the ExoOs group significantly reduced the demyelination area in the corpus callosum compared to the PBS and Exos groups. Additionally, the PDGFRα/APC staining of the demyelinating region revealed an increase in APC+ oligodendrocytes and a decrease in PDGFRα+ oligodendrocyte progenitor cells (OPCs) in the ExoOs group. Furthermore, there was evident myelin regeneration in the demyelinated areas after ExoOs treatment, with better g-ratio and a higher number of intact myelin compared to the other treatment groups. The level of Sox10 expression in the brain tissue of the ExoOs group were higher compared to those of the PBS and Exos groups. The demyelination process can be significantly slowed down by the XPack-modified exosomes, the differentiation of OPCs promoted, and myelin regeneration accelerated under pathological conditions. This process is presumed to be achieved by changing the expression level of intracellular differentiation-related genes after exosomes transport Olig2 enriched into oligodendrocyte progenitors.


Asunto(s)
Cuprizona , Enfermedades Desmielinizantes , Exosomas , Factor de Transcripción 2 de los Oligodendrocitos , Exosomas/metabolismo , Cuprizona/toxicidad , Animales , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/patología , Factor de Transcripción 2 de los Oligodendrocitos/metabolismo , Humanos , Células HEK293 , Vaina de Mielina/metabolismo , Vaina de Mielina/patología , Remielinización/fisiología , Ratones , Células Precursoras de Oligodendrocitos/metabolismo , Ratones Endogámicos C57BL , Cuerpo Calloso/metabolismo , Cuerpo Calloso/patología , Masculino , Oligodendroglía/metabolismo , Modelos Animales de Enfermedad
11.
Glia ; 72(9): 1674-1692, 2024 09.
Artículo en Inglés | MEDLINE | ID: mdl-38899731

RESUMEN

Spinal cord injury (SCI) can result in severe motor and sensory deficits, for which currently no effective cure exists. The pathological process underlying this injury is extremely complex and involves many cell types in the central nervous system. In this study, we have uncovered a novel function for macrophage G protein-coupled receptor kinase-interactor 1 (GIT1) in promoting remyelination and functional repair after SCI. Using GIT1flox/flox Lyz2-Cre (GIT1 CKO) mice, we identified that GIT1 deficiency in macrophages led to an increased generation of tumor necrosis factor-alpha (TNFα), reduced proportion of mature oligodendrocytes (mOLs), impaired remyelination, and compromised functional recovery in vivo. These effects in GIT1 CKO mice were reversed with the administration of soluble TNF inhibitor. Moreover, bone marrow transplantation from GIT1 CWT mice reversed adverse outcomes in GIT1 CKO mice, further indicating the role of macrophage GIT1 in modulating spinal cord injury repair. Our in vitro experiments showed that macrophage GIT1 plays a critical role in secreting TNFα and influences the differentiation of oligodendrocyte precursor cells (OPCs) after stimulation with myelin debris. Collectively, our data uncovered a new role of macrophage GIT1 in regulating the transformation of OPCs into mOLs, essential for functional remyelination after SCI, suggesting that macrophage GIT1 could be a promising treatment target of SCI.


Asunto(s)
Diferenciación Celular , Macrófagos , Células Precursoras de Oligodendrocitos , Remielinización , Traumatismos de la Médula Espinal , Animales , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología , Macrófagos/metabolismo , Remielinización/fisiología , Diferenciación Celular/fisiología , Células Precursoras de Oligodendrocitos/metabolismo , Ratones , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Recuperación de la Función/fisiología , Modelos Animales de Enfermedad , Factor de Necrosis Tumoral alfa/metabolismo , Ratones Transgénicos , Femenino , Proteínas Activadoras de GTPasa/metabolismo , Proteínas Activadoras de GTPasa/genética , Oligodendroglía/metabolismo
12.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167270, 2024 10.
Artículo en Inglés | MEDLINE | ID: mdl-38823461

RESUMEN

Stroke is a major public health concern, with limited clinically approved interventions available to enhance sensorimotor recovery beyond reperfusion. Remarkably, spontaneous recovery is observed in certain stroke patients, suggesting the existence of a brain self-repair mechanism not yet fully understood. In a rat model of permanent cerebral ischemia, we described an increase in oligodendrocytes expressing 3RTau in damaged area. Considering that restoration of myelin integrity ameliorates symptoms in many neurodegenerative diseases, here we hypothesize that this cellular response could trigger remyelination. Our results revealed after ischemia an early recruitment of OPCs to damaged area, followed by their differentiation into 3RTau+ pre-myelinating cells and subsequent into remyelinating oligodendrocytes. Using rat brain slices and mouse primary culture we confirmed the presence of 3RTau in pre-myelinating and a subset of mature oligodendrocytes. The myelin status analysis confirmed long-term remyelination in the damaged area. Postmortem samples from stroke subjects showed a reduction in oligodendrocytes, 3RTau+ cells, and myelin complexity in subcortical white matter. In conclusion, the dynamics of oligodendrocyte populations after ischemia reveals a spontaneous brain self-repair mechanism which restores the functionality of neuronal circuits long-term by remyelination of damaged area. This is evidenced by the improvement of sensorimotor functions in ischemic rats. A deep understanding of this mechanism could be valuable in the search for alternative oligodendrocyte-based, therapeutic interventions to reduce the effects of stroke.


Asunto(s)
Isquemia Encefálica , Vaina de Mielina , Oligodendroglía , Remielinización , Animales , Oligodendroglía/metabolismo , Oligodendroglía/patología , Remielinización/fisiología , Ratas , Vaina de Mielina/metabolismo , Vaina de Mielina/patología , Isquemia Encefálica/patología , Isquemia Encefálica/metabolismo , Humanos , Ratones , Masculino , Diferenciación Celular , Modelos Animales de Enfermedad , Sustancia Blanca/patología , Sustancia Blanca/metabolismo , Femenino , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/metabolismo , Ratones Endogámicos C57BL
13.
Glia ; 72(9): 1555-1571, 2024 09.
Artículo en Inglés | MEDLINE | ID: mdl-38829008

RESUMEN

As one of the top causes of blindness worldwide, glaucoma leads to diverse optic neuropathies such as degeneration of retinal ganglion cells (RGCs). It is widely accepted that the level of intraocular pressure (IOP) is a major risk factor in human glaucoma, and reduction of IOP level is the principally most well-known method to prevent cell death of RGCs. However, clinical studies show that lowering IOP fails to prevent RGC degeneration in the progression of glaucoma. Thus, a comprehensive understanding of glaucoma pathological process is required for developing new therapeutic strategies. In this study, we provide functional and histological evidence showing that optic nerve defects occurred before retina damage in an ocular hypertension glaucoma mouse model, in which oligodendroglial lineage cells were responsible for the subsequent neuropathology. By treatment with clemastine, an Food and Drug Administration (FDA)-approved first-generation antihistamine medicine, we demonstrate that the optic nerve and retina damages were attenuated via promoting oligodendrocyte precursor cell (OPC) differentiation and enhancing remyelination. Taken together, our results reveal the timeline of the optic neuropathies in glaucoma and highlight the potential role of oligodendroglial lineage cells playing in its treatment. Clemastine may be used in future clinical applications for demyelination-associated glaucoma.


Asunto(s)
Clemastina , Glaucoma , Ratones Endogámicos C57BL , Remielinización , Retina , Animales , Clemastina/farmacología , Clemastina/uso terapéutico , Glaucoma/patología , Glaucoma/tratamiento farmacológico , Retina/patología , Retina/efectos de los fármacos , Remielinización/efectos de los fármacos , Remielinización/fisiología , Ratones , Nervio Óptico/efectos de los fármacos , Nervio Óptico/patología , Modelos Animales de Enfermedad , Enfermedades del Nervio Óptico/tratamiento farmacológico , Enfermedades del Nervio Óptico/patología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/patología , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/patología
14.
Neurosci Lett ; 836: 137869, 2024 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-38852766

RESUMEN

Dietary administration of a copper chelator, cuprizone (CPZ), has long been reported to induce intense and reproducible demyelination of several brain structures such as the corpus callosum. Despite the widespread use of CPZ as an animal model for demyelinating diseases such as multiple sclerosis (MS), the mechanism by which it induces demyelination and then allows robust remyelination is still unclear. An intensive mapping of the cell dynamics of oligodendrocyte (OL) lineage during the de- and remyelination course would be particularly important for a deeper understanding of this model. Here, using a panel of OL lineage cell markers as in situ hybridization (ISH) probes, including Pdgfra, Plp, Mbp, Mog, Enpp6, combined with immunofluorescence staining of CC1, SOX10, we provide a detailed dynamic profile of OL lineage cells during the entire course of the model from 1, 2, 3.5 days, 1, 2, 3, 4,5 weeks of CPZ treatment, as well as after 1, 2, 3, 4 weeks of recovery from CPZ treatment. The result showed an unexpected early death of mature OLs and response of OL progenitor cells (OPCs) in vivo upon CPZ challenge, and a prolonged upregulation of myelin-forming OLs compared to the intact control even 4 weeks after CPZ withdrawal. These data may serve as a basic reference system for future studies of the effects of any intervention on de- and remyelination using the CPZ model, and imply the need to optimize the timing windows for the introduction of pro-remyelination therapies in demyelinating diseases such as MS.


Asunto(s)
Linaje de la Célula , Cuprizona , Enfermedades Desmielinizantes , Oligodendroglía , Cuprizona/toxicidad , Animales , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/patología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/patología , Oligodendroglía/metabolismo , Modelos Animales de Enfermedad , Hibridación in Situ/métodos , Ratones Endogámicos C57BL , Ratones , Remielinización/efectos de los fármacos , Remielinización/fisiología , Masculino , Quelantes/toxicidad , Quelantes/farmacología , Vaina de Mielina/patología , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/metabolismo
15.
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
16.
J Neurosci ; 44(28)2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38749703

RESUMEN

Dysregulation of oligodendrocyte progenitor cell (OPC) recruitment and oligodendrocyte differentiation contribute to failure of remyelination in human demyelinating diseases such as multiple sclerosis (MS). Deletion of muscarinic receptor enhances OPC differentiation and remyelination. However, the role of ligand-dependent signaling versus constitutive receptor activation is unknown. We hypothesized that dysregulated acetylcholine (ACh) release upon demyelination contributes to ligand-mediated activation hindering myelin repair. Following chronic cuprizone (CPZ)-induced demyelination (male and female mice), we observed a 2.5-fold increase in ACh concentration. This increase in ACh concentration could be attributed to increased ACh synthesis or decreased acetylcholinesterase-/butyrylcholinesterase (BChE)-mediated degradation. Using choline acetyltransferase (ChAT) reporter mice, we identified increased ChAT-GFP expression following both lysolecithin and CPZ demyelination. ChAT-GFP expression was upregulated in a subset of injured and uninjured axons following intraspinal lysolecithin-induced demyelination. In CPZ-demyelinated corpus callosum, ChAT-GFP was observed in Gfap+ astrocytes and axons indicating the potential for neuronal and astrocytic ACh release. BChE expression was significantly decreased in the corpus callosum following CPZ demyelination. This decrease was due to the loss of myelinating oligodendrocytes which were the primary source of BChE. To determine the role of ligand-mediated muscarinic signaling following lysolecithin injection, we administered neostigmine, a cholinesterase inhibitor, to artificially raise ACh. We identified a dose-dependent decrease in mature oligodendrocyte density with no effect on OPC recruitment. Together, these results support a functional role of ligand-mediated activation of muscarinic receptors following demyelination and suggest that dysregulation of ACh homeostasis directly contributes to failure of remyelination in MS.


Asunto(s)
Enfermedades Desmielinizantes , Oligodendroglía , Transducción de Señal , Animales , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/patología , Ratones , Oligodendroglía/metabolismo , Oligodendroglía/efectos de los fármacos , Femenino , Masculino , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Ratones Endogámicos C57BL , Acetilcolina/metabolismo , Cuprizona/toxicidad , Lisofosfatidilcolinas/toxicidad , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Colina O-Acetiltransferasa/metabolismo , Remielinización/fisiología , Remielinización/efectos de los fármacos , Vaina de Mielina/metabolismo , Vaina de Mielina/efectos de los fármacos , Ratones Transgénicos
17.
Glia ; 72(8): 1469-1483, 2024 08.
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
18.
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
19.
Glia ; 72(8): 1392-1401, 2024 08.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA