Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 628
Filtrar
1.
Nat Commun ; 15(1): 8570, 2024 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-39384784

RESUMEN

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


Asunto(s)
Esclerosis Múltiple , Células Precursoras de Oligodendrocitos , Remielinización , Animales , Remielinización/genética , Humanos , Células Precursoras de Oligodendrocitos/metabolismo , Células Precursoras de Oligodendrocitos/citología , Esclerosis Múltiple/terapia , Esclerosis Múltiple/genética , Sistemas CRISPR-Cas , Células Madre Embrionarias Humanas/metabolismo , Células Madre Embrionarias Humanas/citología , Ratas , Ratones , Edición Génica/métodos , Modelos Animales de Enfermedad , Vaina de Mielina/metabolismo , Femenino , Oligodendroglía/citología , Oligodendroglía/metabolismo , Masculino , Movimiento Celular/genética , Encefalomielitis Autoinmune Experimental/terapia , Encefalomielitis Autoinmune Experimental/genética , Diferenciación Celular
2.
Front Immunol ; 15: 1452172, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39257581

RESUMEN

Background: Glioma is a highly heterogeneous malignancy of the central nervous system. This heterogeneity is driven by various molecular processes, including neoplastic transformation, cell cycle dysregulation, and angiogenesis. Among these biomolecular events, inflammation and stress pathways in the development and driving factors of glioma heterogeneity have been reported. However, the mechanisms of glioma heterogeneity under stress response remain unclear, especially from a spatial aspect. Methods: This study employed single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics (ST) to explore the impact of oxidative stress response genes in oligodendrocyte precursor cells (OPCs). Our analysis identified distinct pathways activated by oxidative stress in two different types of gliomas: high- and low- grade (HG and LG) gliomas. Results: In HG gliomas, oxidative stress induced a metabolic shift from oxidative phosphorylation to glycolysis, promoting cell survival by preventing apoptosis. This metabolic reprogramming was accompanied by epithelial-to-mesenchymal transition (EMT) and an upregulation of stress response genes. Furthermore, SCENIC (Single-Cell rEgulatory Network Inference and Clustering) analysis revealed that oxidative stress activated the AP1 transcription factor in HG gliomas, thereby enhancing tumor cell survival and proliferation. Conclusion: Our findings provide a novel perspective on the mechanisms of oxidative stress responses across various grades of gliomas. This insight enhances our comprehension of the evolutionary processes and heterogeneity within gliomas, potentially guiding future research and therapeutic strategies.


Asunto(s)
Neoplasias Encefálicas , Glioma , Estrés Oxidativo , Análisis de la Célula Individual , Transcriptoma , Glioma/genética , Glioma/patología , Glioma/metabolismo , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/metabolismo , Humanos , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Células Precursoras de Oligodendrocitos/metabolismo , Perfilación de la Expresión Génica , Transducción de Señal , Proliferación Celular/genética , Línea Celular Tumoral , Redes Reguladoras de Genes
3.
Elife ; 132024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39259197

RESUMEN

The emergence of myelinating oligodendrocytes represents a pivotal developmental milestone in vertebrates, given their capacity to ensheath axons and facilitate the swift conduction of action potentials. It is widely accepted that cortical oligodendrocyte progenitor cells (OPCs) arise from medial ganglionic eminence (MGE), lateral/caudal ganglionic eminence (LGE/CGE), and cortical radial glial cells (RGCs). Here, we used two different fate mapping strategies to challenge the established notion that the LGE generates cortical OPCs. Furthermore, we used a Cre/loxP-dependent exclusion strategy to reveal that the LGE/CGE does not give rise to cortical OPCs. Additionally, we showed that specifically eliminating MGE-derived OPCs leads to a significant reduction of cortical OPCs. Together, our findings indicate that the LGE does not generate cortical OPCs, contrary to previous beliefs. These findings provide a new view of the developmental origins of cortical OPCs and a valuable foundation for future research on both normal development and oligodendrocyte-related disease.


Asunto(s)
Corteza Cerebral , Oligodendroglía , Animales , Oligodendroglía/fisiología , Oligodendroglía/citología , Ratones , Corteza Cerebral/embriología , Corteza Cerebral/fisiología , Corteza Cerebral/citología , Células Precursoras de Oligodendrocitos/fisiología , Células Precursoras de Oligodendrocitos/citología , Diferenciación Celular , Eminencia Ganglionar
4.
Int J Med Sci ; 21(11): 2189-2200, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39239553

RESUMEN

In the realm of this study, obtaining a comprehensive understanding of ischemic brain injury and its molecular foundations is of paramount importance. Our study delved into single-cell data analysis, with a specific focus on sub-celltypes and differentially expressed genes in the aftermath of ischemic injury. Notably, we observed a significant enrichment of the "ATP METABOLIC PROCESS" and "ATP HYDROLYSIS ACTIVITY" pathways, featuring pivotal genes such as Pbx3, Dguok, and Kif21b. A remarkable finding was the consistent upregulation of genes like Fabp7 and Bcl11a within the MCAO group, highlighting their crucial roles in regulating the pathway of mitochondrial ATP synthesis coupled proton transport. Furthermore, our network analysis unveiled pathways like "Neuron differentiation" and "T cell differentiation" as central in the regulatory processes of sub-celltypes. These findings provide valuable insights into the intricate molecular responses and regulatory mechanisms that govern brain injury. The shared differentially expressed genes among sub-celltypes emphasize their significance in orchestrating responses post-ischemic injury. Our research, viewed from the perspective of a medical researcher, contributes to the evolving understanding of the molecular landscape underlying ischemic brain injury, potentially paving the way for targeted therapeutic strategies and improved patient outcomes.


Asunto(s)
Adenosina Trifosfato , Infarto de la Arteria Cerebral Media , Cinesinas , Mitocondrias , Células Precursoras de Oligodendrocitos , Transducción de Señal , Animales , Transducción de Señal/genética , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/metabolismo , Mitocondrias/metabolismo , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/biosíntesis , Cinesinas/genética , Cinesinas/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Humanos , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Masculino , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Ratas , Proteínas Proto-Oncogénicas
5.
Sci Rep ; 14(1): 22334, 2024 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-39333617

RESUMEN

The corpus callosum, a major white matter tract in the brain, undergoes age-related functional changes. To extend our investigation of age-related gene expression dynamics in the mouse corpus callosum, we compared RNA-seq data from 2 week-old and 12 week-old wild-type C57BL/6 J mice and identified the differentially expressed genes (e.g., Marcksl1, Chst3, C4b, Neat1, Ndrg1, Emid1, etc.) between these ages. Interestingly, we found that genes highly expressed in myelinating oligodendrocytes were upregulated in 12 week-old mice compared to 2 week-old mice, while genes highly expressed in oligodendrocyte precursor cells (OPCs) and newly formed oligodendrocytes were downregulated. Furthermore, by comparing these genes with the datasets from 20 week-old and 96 week-old mice, we identified novel sets of genes with age-dependent variations in the corpus callosum. These gene expression changes potentially affect key biological pathways and may be closely linked to age-related neurological disorders, including dementia and stroke. Therefore, our results provide an additional dataset to explore age-dependent gene expression dynamics of oligodendrocyte lineage cells in the corpus callosum.


Asunto(s)
Linaje de la Célula , Cuerpo Calloso , Ratones Endogámicos C57BL , Oligodendroglía , Transcriptoma , Animales , Cuerpo Calloso/metabolismo , Cuerpo Calloso/citología , Ratones , Oligodendroglía/metabolismo , Oligodendroglía/citología , Linaje de la Célula/genética , Células Precursoras de Oligodendrocitos/metabolismo , Células Precursoras de Oligodendrocitos/citología , Envejecimiento/genética , Perfilación de la Expresión Génica , Masculino , Factores de Edad
6.
Nat Commun ; 15(1): 7791, 2024 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-39242637

RESUMEN

Multiple sclerosis (MS) is a debilitating demyelinating disease characterized by remyelination failure attributed to inadequate oligodendrocyte precursor cells (OPCs) differentiation and aberrant astrogliosis. A comprehensive cell atlas reanalysis of clinical specimens brings to light heightened clusterin (CLU) expression in a specific astrocyte subtype links to active lesions in MS patients. Our investigation reveals elevated astrocytic CLU levels in both active lesions of patient tissues and female murine MS models. CLU administration stimulates primary astrocyte proliferation while concurrently impeding astrocyte-mediated clearance of myelin debris. Intriguingly, CLU overload directly impedes OPC differentiation and induces OPCs and OLs apoptosis. Mechanistically, CLU suppresses PI3K-AKT signaling in primary OPCs via very low-density lipoprotein receptor. Pharmacological activation of AKT rescues the damage inflicted by excess CLU on OPCs and ameliorates demyelination in the corpus callosum. Furthermore, conditional knockout of CLU emerges as a promising intervention, showcasing improved remyelination processes and reduced severity in murine MS models.


Asunto(s)
Astrocitos , Clusterina , Enfermedades Desmielinizantes , Modelos Animales de Enfermedad , Remielinización , Animales , Femenino , Humanos , Ratones , Apoptosis/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Clusterina/metabolismo , Clusterina/genética , Cuerpo Calloso/metabolismo , Cuerpo Calloso/patología , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Vaina de Mielina/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Células Precursoras de Oligodendrocitos/efectos de los fármacos , Oligodendroglía/metabolismo , Oligodendroglía/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Remielinización/efectos de los fármacos , Transducción de Señal
7.
Int Immunopharmacol ; 142(Pt B): 113117, 2024 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-39293313

RESUMEN

BACKGROUND: The main causes of abnormal white matter development (periventricular leukomalacia) in premature infants are perinatal inflammation and the consequent oxidant/antioxidant imbalance in oligodendrocyte precursor cells (OPCs); however, the underlying mechanisms remain largely unclear. In this work, a rat model of prenatal inflammation was used to examine the mechanism by which artemisinin (ART) protects against white matter dysplasia. METHODS: We established a primary OPC model and rat model of perinatal inflammation. ART was identified from the FDA-approved medicinal chemical library to be beneficial for treating OPC inflammation in model systems. Based on bioinformatics analysis of protein interactions and molecular docking analysis, we further identified the possible targets of ART and evaluated its specific effects and the underlying molecular mechanisms in vivo and in vitro. RESULTS: Following inflammatory stimulation, ART strongly promoted the maturation of OPCs and the development of white matter in the brain. A Cellular thermal shift assay (CETSA) demonstrated that interleukin-1 receptor-associated kinase-4 (IRAK-4) and interleukin-1 receptor-associated kinase-1 (IRAK-1) may be targets of ART, which was consistent with the findings from molecular modelling with Autodock software. Experiments conducted both in vivo and in vitro demonstrated the activation of the IRAK-4/IRAK-1/nuclear factor kappa-B (NF-κB) pathway and the production of inflammatory factors (IL-1ß, IL-6, and TNF-α) in OPCs were greatly suppressed in the group treated with ART compared to the lipopolysaccharide (LPS)-treated group. Moreover, ART dramatically decreased reactive oxygen species (ROS) levels in OPCs while increasing nuclear factor e2-related factor 2 (Nrf2) levels. CONCLUSION: Our findings suggest that ART can significantly reduce OPC perinatal inflammation and consequent oxidative stress. The targeted inhibition of IRAK-4 and IRAK-1 by ART may be a potential therapeutic strategy for alleviating abnormalities in white matter development in premature newborns.


Asunto(s)
Artemisininas , Quinasas Asociadas a Receptores de Interleucina-1 , Células Precursoras de Oligodendrocitos , Estrés Oxidativo , Animales , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Estrés Oxidativo/efectos de los fármacos , Células Precursoras de Oligodendrocitos/efectos de los fármacos , Células Precursoras de Oligodendrocitos/metabolismo , Ratas , Femenino , Artemisininas/farmacología , Artemisininas/uso terapéutico , Embarazo , Humanos , Inflamación/tratamiento farmacológico , Células Cultivadas , Ratas Sprague-Dawley , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Simulación del Acoplamiento Molecular , Modelos Animales de Enfermedad , Animales Recién Nacidos , FN-kappa B/metabolismo
8.
J Cell Biol ; 223(11)2024 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-39133301

RESUMEN

Adult oligodendrocyte progenitors (aOPCs) generate myelinating oligodendrocytes like neonatal progenitors (nOPCs), and they also display unique functional features. Here, using unbiased histone proteomics analysis and ChIP sequencing analysis of PDGFRα+ OPCs sorted from neonatal and adult Pdgfra-H2B-EGFP reporter mice, we identify the activating H4K8ac histone mark as enriched in the aOPCs. We detect increased occupancy of the H4K8ac activating mark at chromatin locations corresponding to genes related to the progenitor state (e.g., Hes5, Gpr17), metabolic processes (e.g., Txnip, Ptdgs), and myelin components (e.g., Cnp, Mog). aOPCs showed higher levels of transcripts related to lipid metabolism and myelin, and lower levels of transcripts related to cell cycle and proliferation compared with nOPCs. In addition, pharmacological inhibition of histone acetylation decreased the expression of the H4K8ac target genes in aOPCs and decreased their proliferation. Overall, this study identifies acetylation of the histone H4K8 as a regulator of the proliferative capacity of aOPCs.


Asunto(s)
Proliferación Celular , Histonas , Células Precursoras de Oligodendrocitos , Animales , Histonas/metabolismo , Histonas/genética , Acetilación , Células Precursoras de Oligodendrocitos/metabolismo , Células Precursoras de Oligodendrocitos/citología , Ratones , Oligodendroglía/metabolismo , Oligodendroglía/citología , Diferenciación Celular , Células Cultivadas , Ratones Endogámicos C57BL
9.
Neoplasia ; 57: 101042, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39216363

RESUMEN

Proneural gliomas are brain tumors characterized by enrichment of oligodendrocyte progenitor cell (OPC) transcripts and genetic alterations. In this study we sought to identify transcriptional and epigenetic differences between OPCs with Trp53 deletion and PDGF-BB overexpression (BB-p53n) and those carrying only p53 deletion (p53n). In culture, the BB-p53n OPCs display growth characteristics more similar to glioma cells than p53n OPCs. When injected in mouse brains, BB-p53n OPC form tumors, while the p53n OPCs do not. Unbiased histone proteomics and transcriptomic analysis on these OPC populations identified higher levels of the histone H3K27me3 mark and lower levels of the histone H4K20me3. The transcriptome of the BB-p53n OPCs was characterized by higher levels of transcripts related to proliferation and cell adhesion compared to p53n OPCs. Pharmacological inhibition of the enzyme responsible for histone H3K27 trimethylation (EZH2i) in BB-p53n OPCs, reduced cell cycle transcripts and increased the expression of differentiation markers, but was not sufficient to restore their growth characteristics. This suggests that PDGF-BB overexpression in p53n OPCs favors the early stages of transformation, by promoting proliferation and halting differentiation in a H3K27me3-dependent pathway, and favoring growth characteristics in a H3K27me3 independent manner.


Asunto(s)
Becaplermina , Proliferación Celular , Histonas , Células Precursoras de Oligodendrocitos , Proteína p53 Supresora de Tumor , Animales , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ratones , Histonas/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Becaplermina/metabolismo , Becaplermina/genética , Diferenciación Celular/genética , Metilación , Humanos , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Epigénesis Genética
10.
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
11.
Mol Brain ; 17(1): 56, 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39138468

RESUMEN

Several prior studies have proposed the involvement of various brain regions and cell types in Parkinson's disease (PD) pathology. Here, we performed snRNA-seq on the prefrontal cortex and anterior cingulate regions from a small cohort of post-mortem control and PD brain tissue. We found a significant association of oligodendrocytes (ODCs) and oligodendrocyte precursor cells (OPCs) with PD-linked risk loci and report several dysregulated genes and pathways, including regulation of tau-protein kinase activity, regulation of inclusion body assembly and protein processing involved in protein targeting to mitochondria. In an independent PD cohort with clinical measures (681 cases and 549 controls), polygenic risk scores derived from the dysregulated genes significantly predicted Montreal Cognitive Assessment (MoCA)-, and Beck Depression Inventory-II (BDI-II)-scores but not motor impairment (UPDRS-III). We extended our analysis of clinical outcome prediction by incorporating differentially expressed genes from three separate datasets that were previously published by different laboratories. In the first dataset from the anterior cingulate cortex, we identified an association between ODCs and BDI-II. In the second dataset obtained from the substantia nigra (SN), OPCs displayed an association with UPDRS-III. In the third dataset from the SN region, a distinct subtype of OPCs, labeled OPC_ADM, exhibited an association with UPDRS-III. Intriguingly, the OPC_ADM cluster also demonstrated a significant increase in PD samples. These results suggest that by expanding our focus to glial cells, we can uncover region-specific molecular pathways associated with PD symptoms.


Asunto(s)
Oligodendroglía , Enfermedad de Parkinson , Transcriptoma , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Humanos , Oligodendroglía/metabolismo , Oligodendroglía/patología , Transcriptoma/genética , Masculino , Femenino , Anciano , Células Precursoras de Oligodendrocitos/metabolismo , Estudios de Cohortes , Persona de Mediana Edad
12.
Nat Commun ; 15(1): 6979, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-39143079

RESUMEN

Oligodendrocyte precursor cells (OPCs) give rise to myelinating oligodendrocytes of the brain. This process persists throughout life and is essential for recovery from neurodegeneration. To better understand the cellular checkpoints that occur during oligodendrogenesis, we determined the mitochondrial distribution and morphometrics across the oligodendrocyte lineage in mouse and human cerebral cortex. During oligodendrocyte generation, mitochondrial content expands concurrently with a change in subcellular partitioning towards the distal processes. These changes are followed by an abrupt loss of mitochondria in the oligodendrocyte processes and myelin, coinciding with sheath compaction. This reorganization and extensive expansion and depletion take 3 days. Oligodendrocyte mitochondria are stationary over days while OPC mitochondrial motility is modulated by animal arousal state within minutes. Aged OPCs also display decreased mitochondrial size, volume fraction, and motility. Thus, mitochondrial dynamics are linked to oligodendrocyte generation, dynamically modified by their local microenvironment, and altered in the aging brain.


Asunto(s)
Mitocondrias , Vaina de Mielina , Oligodendroglía , Animales , Mitocondrias/metabolismo , Humanos , Oligodendroglía/metabolismo , Oligodendroglía/citología , Ratones , Vaina de Mielina/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Células Precursoras de Oligodendrocitos/citología , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Ratones Endogámicos C57BL , Masculino , Dinámicas Mitocondriales , Diferenciación Celular , Femenino
13.
CNS Neurosci Ther ; 30(8): e14925, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39161089

RESUMEN

AIMS: Hypoperfusion induces significant white matter injury in cerebral vascular disorders, including arteriosclerotic cerebral small vessel disease (aCSVD), which is prevalent among the elderly. Iron transport by blood vessel endothelial cells (BVECs) from the periphery supports oligodendrocyte maturation and white matter repair. This study aims to elucidate the association between iron homeostasis changes and white matter injury severity, and explore the crosstalk between BVECs and oligodendroglial lineage cells. METHODS: In vivo: C57BL/6 mice were subjected to unilateral common carotid artery occlusion (UCCAO). In vitro: BVECs with myelin pretreatment were co-cultured with oligodendrocyte progenitor cells (OPCs) or organotypic cerebellar slices subjected to oxygen and glucose deprivation. RESULTS: Circulatory iron tends to be stored in aCSVD patients with white matter injury. Myelin debris endocytosis by BVECs impairs iron transport, trapping iron in the blood and away from the brain, worsening oligodendrocyte iron deficiency in hypoperfusion-induced white matter injury. Iron accumulation in BVECs triggers ferroptosis, suppressing iron transport and hindering white matter regeneration. Intranasal holo-transferrin (hTF) administration bypassing the BBB alleviates oligodendrocyte iron deficiency and promotes myelin regeneration in hypoperfusion-induced white matter injury. CONCLUSION: The iron imbalance between BVECs and oligodendroglial lineage cells is a potential therapeutic target in hypoperfusion-induced white matter injury.


Asunto(s)
Endocitosis , Células Endoteliales , Hierro , Ratones Endogámicos C57BL , Vaina de Mielina , Oligodendroglía , Sustancia Blanca , Animales , Células Endoteliales/metabolismo , Células Endoteliales/patología , Ratones , Oligodendroglía/metabolismo , Oligodendroglía/patología , Sustancia Blanca/metabolismo , Sustancia Blanca/patología , Hierro/metabolismo , Vaina de Mielina/metabolismo , Vaina de Mielina/patología , Endocitosis/fisiología , Endocitosis/efectos de los fármacos , Masculino , Sobrecarga de Hierro/metabolismo , Sobrecarga de Hierro/patología , Encéfalo/metabolismo , Encéfalo/patología , Células Precursoras de Oligodendrocitos/metabolismo , Células Precursoras de Oligodendrocitos/efectos de los fármacos , Células Precursoras de Oligodendrocitos/patología
14.
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
15.
Neurosci Bull ; 40(10): 1573-1589, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39042298

RESUMEN

Oligodendrocyte precursor cells (OPCs) are a heterogeneous multipotent population in the central nervous system (CNS) that appear during embryogenesis and persist as resident cells in the adult brain parenchyma. OPCs could generate oligodendrocytes to participate in myelination. Recent advances have renewed our knowledge of OPC biology by discovering novel markers of oligodendroglial cells, the myelin-independent roles of OPCs, and the regulatory mechanism of OPC development. In this review, we will explore the updated knowledge on OPC identity, their multifaceted roles in the CNS in health and diseases, as well as the regulatory mechanisms that are involved in their developmental stages, which hopefully would contribute to a further understanding of OPCs and attract attention in the field of OPC biology.


Asunto(s)
Sistema Nervioso Central , Células Precursoras de Oligodendrocitos , Animales , Células Precursoras de Oligodendrocitos/fisiología , Células Precursoras de Oligodendrocitos/citología , Humanos , Sistema Nervioso Central/citología , Sistema Nervioso Central/fisiología , Oligodendroglía/fisiología , Diferenciación Celular/fisiología , Vaina de Mielina/fisiología
16.
Front Immunol ; 15: 1425706, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39044821

RESUMEN

Oligodendrocyte precursor cells (OPCs) have long been regarded as progenitors of oligodendrocytes, yet recent advances have illuminated their multifaceted nature including their emerging immune functions. This review seeks to shed light on the immune functions exhibited by OPCs, spanning from phagocytosis to immune modulation and direct engagement with immune cells across various pathological scenarios. Comprehensive understanding of the immune functions of OPCs alongside their other roles will pave the way for targeted therapies in neurological disorders.


Asunto(s)
Células Precursoras de Oligodendrocitos , Humanos , Células Precursoras de Oligodendrocitos/inmunología , Animales , Fagocitosis/inmunología , Oligodendroglía/inmunología , Diferenciación Celular/inmunología , Inmunomodulación
17.
PLoS Biol ; 22(7): e3002655, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38985832

RESUMEN

Oligodendrocyte precursor cells (OPCs) are a class of glial cells that uniformly tiles the entire central nervous system (CNS). They play several key functions across the brain including the generation of oligodendrocytes and the control of myelination. Whether the functional diversity of OPCs is the result of genetically defined subpopulations or of their regulation by external factors has not been definitely established. We discovered that a subpopulation of OPCs found across the brain is defined by the expression of C1ql1, a gene previously described for its synaptic function in neurons. This subpopulation starts to appear during the first postnatal week in the mouse cortex. Ablation of C1ql1-expressing OPCs in the mouse leads to a massive lack of oligodendrocytes and myelination in many brain regions. This deficit cannot be rescued, even though some OPCs escape Sox10-driven ablation and end up partially compensating the OPC loss in the adult. Therefore, C1ql1 is a molecular marker of a functionally non-redundant subpopulation of OPCs, which controls the generation of myelinating oligodendrocytes.


Asunto(s)
Vaina de Mielina , Células Precursoras de Oligodendrocitos , Oligodendroglía , Animales , Células Precursoras de Oligodendrocitos/metabolismo , Células Precursoras de Oligodendrocitos/citología , Oligodendroglía/metabolismo , Oligodendroglía/citología , Vaina de Mielina/metabolismo , Ratones , Diferenciación Celular/genética , Encéfalo/metabolismo , Encéfalo/citología , Encéfalo/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica
18.
PLoS Biol ; 22(7): e3002691, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38990827

RESUMEN

The diversity of oligodendrocyte precursor cells (OPCs) is not well understood and is actively discussed in the field. A new study in PLOS Biology describes a novel marker for an OPC subpopulation that controls oligodendrogenesis and myelination.


Asunto(s)
Diferenciación Celular , Oligodendroglía , Oligodendroglía/fisiología , Oligodendroglía/metabolismo , Oligodendroglía/citología , Animales , Humanos , Vaina de Mielina/metabolismo , Vaina de Mielina/fisiología , Células Precursoras de Oligodendrocitos/fisiología , Células Precursoras de Oligodendrocitos/citología , Células Precursoras de Oligodendrocitos/metabolismo , Biomarcadores/metabolismo
19.
Cells ; 13(13)2024 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-38994940

RESUMEN

The abnormal growth of oligodendrocyte precursor cells (OPCs) significantly contributes to the progression of glioblastoma tumors. Hence, molecules that block OPC growth may be of therapeutic importance in treating gliomas. 2-Methoxyestradiol (2ME), an endogenous tubulin-interacting metabolite of estradiol, is effective against multiple proliferative disorders. Based on its anti-carcinogenic and anti-angiogenic actions, it is undergoing phase II clinical trials. We hypothesize that 2ME may prevent glioma growth by targeting OPC growth. Here, we tested this hypothesis by assessing the impact of 2ME on the growth of an OPC line, "Oli-neu", and dissected the underlying mechanism(s). Treatment with 2ME inhibited OPC growth in a concentration-dependent manner, accompanied by significant upregulation in the expression of p21 and p27, which are negative cell-cycle regulators. Moreover, treatment with 2ME altered OPC morphology from multi-arm processes to rounded cells. At concentrations of 1uM and greater, 2ME induced apoptosis, with increased expressions of caspase 3, PARP, and caspase-7 fragments, externalized phosphatidylserine staining/APOPercentage, and increased mitochondrial activity. Flow cytometry and microscopic analysis demonstrated that 2ME triggers endoreduplication in a concentration-dependent fashion. Importantly, 2ME induced cyclin E, JNK1/2, and p53 expression, as well as OPC fusion, which are key mechanisms driving endoreduplication and whole-genome duplication. Importantly, the inhibition of p53 with pifithrin-α rescued 2ME-induced endoreduplication. The pro-apoptotic and endoreduplication actions of 2ME were accompanied by the upregulation of survivin, cyclin A, Cyclin B, Cyclin D2, and ppRB. Similar growth inhibitory, apoptotic, and endoreduplication effects of 2ME were observed in CG4 cells. Taken together, our findings provide evidence that 2ME not only inhibits OPC growth and triggers apoptosis, but also activates OPCs into survival (fight or flight) mode, leading to endoreduplication. This inherent survival characteristic of OPCs may, in part, be responsible for drug resistance in gliomas, as observed for many tubulin-interacting drugs. Importantly, the fate of OPCs after 2ME treatment may depend on the cell-cycle status of individual cells. Combining tubulin-interfering molecules with drugs such as pifithrin-α that inhibit endoreduplication may help inhibit OPC/glioma growth and limit drug resistance.


Asunto(s)
2-Metoxiestradiol , Apoptosis , Proteína p53 Supresora de Tumor , 2-Metoxiestradiol/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Animales , Células Precursoras de Oligodendrocitos/metabolismo , Células Precursoras de Oligodendrocitos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Humanos , Estradiol/farmacología , Estradiol/análogos & derivados , Oligodendroglía/metabolismo , Oligodendroglía/efectos de los fármacos , Antimitóticos/farmacología , Línea Celular
20.
Sci Adv ; 10(28): eadk9918, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-38996029

RESUMEN

Cell therapy for the treatment of demyelinating diseases such as multiple sclerosis is hampered by poor survival of donor oligodendrocyte cell preparations, resulting in limited therapeutic outcomes. Excessive cell death leads to the release of intracellular alloantigens, which likely exacerbate local inflammation and may predispose the graft to eventual rejection. Here, we engineered innovative cell-instructive shear-thinning hydrogels (STHs) with tunable viscoelasticity and bioactivity for minimally invasive delivery of primary human oligodendrocyte progenitor cells (hOPCs) to the brain of a shiverer/rag2 mouse, a model of congenital hypomyelinating disease. The STHs enabled immobilization of prosurvival signals, including a recombinantly designed bidomain peptide and platelet-derived growth factor. Notably, STHs reduced the death rate of hOPCs significantly, promoted the production of myelinating oligodendrocytes, and enhanced myelination of the mouse brain 12 weeks post-implantation. Our results demonstrate the potential of STHs loaded with biological cues to improve cell therapies for the treatment of devastating myelopathies.


Asunto(s)
Supervivencia Celular , Hidrogeles , Células Precursoras de Oligodendrocitos , Remielinización , Animales , Hidrogeles/química , Células Precursoras de Oligodendrocitos/metabolismo , Células Precursoras de Oligodendrocitos/citología , Ratones , Humanos , Sistema Nervioso Central/metabolismo , Oligodendroglía/metabolismo , Oligodendroglía/citología , Vaina de Mielina/metabolismo , Modelos Animales de Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA