Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Cell ; 187(8): 1955-1970.e23, 2024 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-38503282

RESUMO

Characterizing somatic mutations in the brain is important for disentangling the complex mechanisms of aging, yet little is known about mutational patterns in different brain cell types. Here, we performed whole-genome sequencing (WGS) of 86 single oligodendrocytes, 20 mixed glia, and 56 single neurons from neurotypical individuals spanning 0.4-104 years of age and identified >92,000 somatic single-nucleotide variants (sSNVs) and small insertions/deletions (indels). Although both cell types accumulate somatic mutations linearly with age, oligodendrocytes accumulated sSNVs 81% faster than neurons and indels 28% slower than neurons. Correlation of mutations with single-nucleus RNA profiles and chromatin accessibility from the same brains revealed that oligodendrocyte mutations are enriched in inactive genomic regions and are distributed across the genome similarly to mutations in brain cancers. In contrast, neuronal mutations are enriched in open, transcriptionally active chromatin. These stark differences suggest an assortment of active mutagenic processes in oligodendrocytes and neurons.


Assuntos
Envelhecimento , Encéfalo , Neurônios , Oligodendroglia , Humanos , Envelhecimento/genética , Envelhecimento/patologia , Cromatina/genética , Cromatina/metabolismo , Mutação , Neurônios/metabolismo , Neurônios/patologia , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Análise da Expressão Gênica de Célula Única , Sequenciamento Completo do Genoma , Encéfalo/metabolismo , Encéfalo/patologia , Polimorfismo de Nucleotídeo Único , Mutação INDEL , Bancos de Espécimes Biológicos , Células Precursoras de Oligodendrócitos/metabolismo , Células Precursoras de Oligodendrócitos/patologia
2.
Stem Cell Res Ther ; 15(1): 35, 2024 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-38321505

RESUMO

BACKGROUND: Spinal cord injury (SCI) is a devastating disease that causes extensive damage to oligodendrocytes and neurons leading to demyelination and axonal degeneration. In this study, we co-transplanted cell grafts containing oligodendrocyte progenitor cells (OPCs) derived from human-induced pluripotent stem cells (iPSCs) combined with human umbilical vein endothelial cells (HUVECs), which were reported to promote OPCs survival and migration, into rat contusion models to promote functional recovery after SCI. METHODS: OPCs were derived from iPSCs and identified by immunofluorescence at different time points. Functional assays in vitro were performed to evaluate the effect of HUVECs on the proliferation, migration, and survival of OPCs by co-culture and migration assay, as well as on the neuronal axonal growth. A combination of OPCs and HUVECs was transplanted into the rat contusive model. Upon 8 weeks, immunofluorescence staining was performed to test the safety of transplanted cells and to observe the neuronal repairment, myelination, and neural circuit reconstruction at the injured area; also, the functional recovery was assessed by Basso, Beattie, and Bresnahan open-field scale, Ladder climb, SEP, and MEP. Furthermore, the effect of HUVECs on grafts was also determined in vivo. RESULTS: Data showed that HUVECs promote the proliferation, migration, and survival of OPCs both in vitro and in vivo. Furthermore, 8 weeks upon engraftment, the rats with OPCs and HUVECs co-transplantation noticeably facilitated remyelination, enhanced functional connection between the grafts and the host and promoted functional recovery. In addition, compared with the OPCs-alone transplantation, the co-transplantation generated more sensory neurons at the lesion border and significantly improved the sensory functional recovery. CONCLUSIONS: Our study demonstrates that transplantation of OPCs combined with HUVECs significantly enhances both motor and sensory functional recovery after SCI. No significance was observed between OPCs combined with HUVECs group and OPCs-alone group in motor function recovery, while the sensory function recovery was significantly promoted in OPCs combined with HUVECs groups compared with the other two groups. These findings provide novel insights into the field of SCI research.


Assuntos
Células-Tronco Pluripotentes Induzidas , Células Precursoras de Oligodendrócitos , Traumatismos da Medula Espinal , Ratos , Humanos , Animais , Células Precursoras de Oligodendrócitos/patologia , Células Precursoras de Oligodendrócitos/transplante , Células Endoteliais da Veia Umbilical Humana , Recuperação de Função Fisiológica , Células-Tronco Pluripotentes Induzidas/transplante , Traumatismos da Medula Espinal/patologia , Oligodendroglia , Medula Espinal/patologia , Diferenciação Celular/fisiologia
3.
Ther Hypothermia Temp Manag ; 13(3): 134-140, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36862528

RESUMO

Periventricular leukomalacia (PVL), characterized by distinctive form of white matter injury, often arises after neonatal cardiac surgery. Proven therapies for PVL are absent. In this study, we designed to quest therapeutic effects of delayed mild hypothermia on PVL and its mechanism in a neonatal rat brain slice model. With the increase of delayed mild hypothermia-treating time, the reduced expression of myelin basic protein and loss of preoligodendrocytes were significantly attenuated after oxygen-glucose deprivation. In addition, the proportion of ionized calcium binding adapter molecule 1 (Iba-1)-positive cells and the expression of Iba-1 were apparently reduced with the increased duration of mild hypothermia treatment. Furthermore, the levels of tumor necrosis factor alpha and interleukin-6 reduced after the mild hypothermia treatment relative to the control. Inhibition of microglial activation with prolonged mild hypothermia may be a potential strategy for white matter protection during cardiopulmonary bypass and hypothermic circulatory arrest.


Assuntos
Hipotermia Induzida , Hipotermia , Leucomalácia Periventricular , Células Precursoras de Oligodendrócitos , Ratos , Animais , Animais Recém-Nascidos , Células Precursoras de Oligodendrócitos/metabolismo , Células Precursoras de Oligodendrócitos/patologia , Microglia/metabolismo , Microglia/patologia , Hipotermia/metabolismo , Leucomalácia Periventricular/terapia , Leucomalácia Periventricular/metabolismo , Leucomalácia Periventricular/patologia , Encéfalo/patologia
4.
Front Med ; 17(2): 240-262, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36645634

RESUMO

Detailed characterizations of genomic alterations have not identified subtype-specific vulnerabilities in adult gliomas. Mapping gliomas into developmental programs may uncover new vulnerabilities that are not strictly related to genomic alterations. After identifying conserved gene modules co-expressed with EGFR or PDGFRA (EM or PM), we recently proposed an EM/PM classification scheme for adult gliomas in a histological subtype- and grade-independent manner. By using cohorts of bulk samples, paired primary and recurrent samples, multi-region samples from the same glioma, single-cell RNA-seq samples, and clinical samples, we here demonstrate the temporal and spatial stability of the EM and PM subtypes. The EM and PM subtypes, which progress in a subtype-specific mode, are robustly maintained in paired longitudinal samples. Elevated activities of cell proliferation, genomic instability and microenvironment, rather than subtype switching, mark recurrent gliomas. Within individual gliomas, the EM/PM subtype was preserved across regions and single cells. Malignant cells in the EM and PM gliomas were correlated to neural stem cell and oligodendrocyte progenitor cell compartment, respectively. Thus, while genetic makeup may change during progression and/or within different tumor areas, adult gliomas evolve within a neurodevelopmental framework of the EM and PM molecular subtypes. The dysregulated developmental pathways embedded in these molecular subtypes may contain subtype-specific vulnerabilities.


Assuntos
Neoplasias Encefálicas , Glioma , Células-Tronco Neurais , Células Precursoras de Oligodendrócitos , Humanos , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Recidiva Local de Neoplasia/metabolismo , Glioma/genética , Glioma/metabolismo , Glioma/patologia , Células-Tronco Neurais/patologia , Células Precursoras de Oligodendrócitos/patologia , Microambiente Tumoral
5.
Glia ; 70(9): 1681-1698, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35524725

RESUMO

Diffuse midline glioma (DMG) is a type of lethal brain tumor that develops mainly in children. The majority of DMG harbor the K27M mutation in histone H3. Oligodendrocyte progenitor cells (OPCs) in the brainstem are candidate cells-of-origin for DMG, yet there is no genetically engineered mouse model of DMG initiated in OPCs. Here, we used the RCAS/Tv-a avian retroviral system to generate DMG in Olig2-expressing progenitors and Nestin-expressing progenitors in the neonatal mouse brainstem. PDGF-A or PDGF-B overexpression, along with p53 deletion, resulted in gliomas in both models. Exogenous overexpression of H3.3K27M had a significant effect on tumor latency and tumor cell proliferation when compared with H3.3WT in Nestin+ cells but not in Olig2+ cells. Further, the fraction of H3.3K27M-positive cells was significantly lower in DMGs initiated in Olig2+ cells relative to Nestin+ cells, both in PDGF-A and PDGF-B-driven models, suggesting that the requirement for H3.3K27M is reduced when tumorigenesis is initiated in Olig2+ cells. RNA-sequencing analysis revealed that the differentially expressed genes in H3.3K27M tumors were non-overlapping between Olig2;PDGF-B, Olig2;PDGF-A, and Nestin;PDGF-A models. GSEA analysis of PDGFA tumors confirmed that the transcriptomal effects of H3.3K27M are cell-of-origin dependent with H3.3K27M promoting epithelial-to-mesenchymal transition (EMT) and angiogenesis when Olig2 marks the cell-of-origin and inhibiting EMT and angiogenesis when Nestin marks the cell-of-origin. We did observe some overlap with H3.3K27M promoting negative enrichment of TNFA_Signaling_Via_NFKB in both models. Our study suggests that the tumorigenic effects of H3.3K27M are cell-of-origin dependent, with H3.3K27M being more oncogenic in Nestin+ cells than Olig2+ cells.


Assuntos
Neoplasias Encefálicas , Glioma , Células Precursoras de Oligodendrócitos , Animais , Neoplasias Encefálicas/patologia , Modelos Animais de Doenças , Glioma/patologia , Histonas , Camundongos , Mutação/genética , Nestina/genética , Células Precursoras de Oligodendrócitos/patologia
6.
J Neurosci ; 41(48): 9872-9890, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34725188

RESUMO

Oligodendrocytes are vulnerable to genetic and environmental insults and its injury leads to demyelinating diseases. The roles of ErbB receptors in maintaining the CNS myelin integrity are largely unknown. Here, we overactivate ErbB receptors that mediate signaling of either neuregulin (NRG) or epidermal growth factor (EGF) family growth factors and found their synergistic activation caused deleterious outcomes in white matter. Sustained ErbB activation induced by the tetracycline-dependent mouse tool Plp-tTA resulted in demyelination, axonal degeneration, oligodendrocyte precursor cell (OPC) proliferation, astrogliosis, and microgliosis in white matter. Moreover, there was hypermyelination before these inflammatory pathologic events. In contrast, sustained ErbB activation induced by another tetracycline-dependent mouse tool Sox10+/rtTA caused hypomyelination in the corpus callosum and optic nerve, which appeared to be a developmental deficit and did not associate with OPC regeneration, astrogliosis, or microgliosis. By tracing the differentiation states of cells expressing tetracycline-controlled transcriptional activator (tTA)/reverse tTA (rtTA)-dependent transgene or pulse-labeled reporter proteins in vitro and in vivo, we found that Plp-tTA targeted mainly mature oligodendrocytes (MOs), whereas Sox10+/rtTA targeted OPCs and newly-formed oligodendrocytes (NFOs). The distinct phenotypes of mice with ErbB overactivation induced by Plp-tTA and Sox10+/rtTA consolidated their nonoverlapping targeting preferences in the oligodendrocyte lineage, and enabled us to demonstrate that ErbB overactivation in MOs induced necroptosis that caused inflammatory demyelination, whereas in OPCs induced apoptosis that caused noninflammatory hypomyelination. Early interference with aberrant ErbB activation ceased oligodendrocyte deaths and restored myelin development in both mice. This study suggests that aberrant ErbB activation is an upstream pathogenetic mechanism of demyelinating diseases, providing a potential therapeutic target.SIGNIFICANCE STATEMENT Primary oligodendropathy is one of the etiologic mechanisms for multiple sclerosis, and oligodendrocyte necroptosis is a pathologic hallmark in the disease. Moreover, the demyelinating disease is now a broad concept that embraces schizophrenia, in which white matter lesions are an emerging feature. ErbB overactivation has been implicated in schizophrenia by genetic analysis and postmortem studies. This study suggests the etiologic implications of ErbB overactivation in myelin pathogenesis and elucidates the pathogenetic mechanisms.


Assuntos
Doenças Desmielinizantes/patologia , Receptores ErbB/metabolismo , Neurogênese/fisiologia , Células Precursoras de Oligodendrócitos/patologia , Oligodendroglia/patologia , Animais , Apoptose/fisiologia , Doenças Desmielinizantes/metabolismo , Feminino , Masculino , Camundongos , Necroptose/fisiologia , Oligodendroglia/metabolismo , Substância Branca/patologia
7.
Neuropathology ; 41(3): 161-173, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33913208

RESUMO

Oligodendrocyte precursor cells (OPCs) are a fourth resident glial cell population in the mammalian central nervous system. They are evenly distributed throughout the gray and white matter and continue to proliferate and generate new oligodendrocytes (OLs) throughout life. They were understudied until a few decades ago when immunolabeling for NG2 and platelet-derived growth factor receptor alpha revealed cells that are distinct from mature OLs, astrocytes, neurons, and microglia. In this review, we provide a summary of the known properties of OPCs with some historical background, followed by highlights from recent studies that suggest new roles for OPCs in certain pathological conditions.


Assuntos
Células Precursoras de Oligodendrócitos/patologia , Células Precursoras de Oligodendrócitos/fisiologia , Animais , Antígenos/análise , Antígenos/metabolismo , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Glioma/genética , Humanos , Microscopia Eletrônica , Neurônios , Células Precursoras de Oligodendrócitos/metabolismo , Células Precursoras de Oligodendrócitos/ultraestrutura , Oligodendroglia/fisiologia , Proteoglicanas/análise , Proteoglicanas/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
8.
Biol Pharm Bull ; 44(2): 181-187, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33518671

RESUMO

Oligodendrocyte precursor cells (OPCs) are glial cells that differentiate into oligodendrocytes and myelinate axons. The number of OPCs is reportedly increased in brain lesions in some demyelinating diseases and during ischemia; however, these cells also secrete cytokines and elicit both protective and deleterious effects in response to brain injury. The mechanism regulating the behaviors of OPCs in physiological and pathological conditions must be elucidated to control these cells and to treat demyelinating diseases. Here, we focused on transient receptor potential melastatin 3 (TRPM3), a Ca2+-permeable channel that is activated by the neurosteroid pregnenolone sulfate (PS) and body temperature. Trpm3+/Pdgfra+ OPCs were detected in the cerebral cortex (CTX) and corpus callosum (CC) of P4 and adult rats by in situ hybridization. Trpm3 expression was detected in primary cultured rat OPCs and was increased by treatment with tumor necrosis factor α (TNFα). Application of PS (30-100 µM) increased the Ca2+ concentration in OPCs and this effect was inhibited by co-treatment with the TRP channel blocker Gd3+ (100 µM) or the TRPM3 inhibitor isosakuranetin (10 µM). Stimulation of TRPM3 with PS (50 µM) did not affect the differentiation or migration of OPCs. The number of Trpm3+ OPCs was markedly increased in demyelinated lesions in an endothelin-1 (ET-1)-induced ischemic rat model. In conclusion, TRPM3 is functionally expressed in OPCs in vivo and in vitro and is upregulated in inflammatory conditions such as ischemic insults and TNFα treatment, implying that TRPM3 is involved in the regulation of specific behaviors of OPCs in pathological conditions.


Assuntos
Córtex Cerebral/patologia , Doenças Desmielinizantes/patologia , Células Precursoras de Oligodendrócitos/patologia , Acidente Vascular Cerebral Lacunar/patologia , Canais de Cátion TRPM/metabolismo , Animais , Células Cultivadas , Córtex Cerebral/irrigação sanguínea , Córtex Cerebral/citologia , Corpo Caloso/irrigação sanguínea , Corpo Caloso/citologia , Corpo Caloso/patologia , Doenças Desmielinizantes/etiologia , Modelos Animais de Doenças , Humanos , Células Precursoras de Oligodendrócitos/efeitos dos fármacos , Pregnenolona/farmacologia , Cultura Primária de Células , Ratos , Receptor alfa de Fator de Crescimento Derivado de Plaquetas , Acidente Vascular Cerebral Lacunar/complicações , Canais de Cátion TRPM/agonistas , Regulação para Cima
9.
Exp Biol Med (Maywood) ; 246(10): 1198-1209, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33557607

RESUMO

White matter lesion (WML) is caused by chronic cerebral hypoperfusion, which are usually associated with cognitive impairment. Evidence from recent studies has shown that ginkgolide B has a neuroprotective effect that could be beneficial for the treatment of ischemia; however, it is not clear whether ginkgolide B has a protective effect on WML. Our data show that ginkgolide B can promote the differentiation of oligodendrocyte precursor cell (OPC) into oligodendrocytes and promote oligodendrocyte survival following a WML. Ginkgolide B (5, 10, 20 mg/kg) or saline is administered intraperitoneally every day after WML. After 4 weeks, the data of Morris water maze suggested that rats' memory and learning abilities were impaired, and the administration of ginkgolide B enhanced behavioral achievement. Also, treatment with ginkgolide B significantly attenuated this loss of myelin. Our result suggests that ginkgolide B promotes the differentiation of OPC into oligodendrocytes. We also found that ginkgolide B ameliorates oligodendrocytes apoptosis. Furthermore, ginkgolide B enhanced the expression of phosphorylated Akt and CREB. In conclusion, our data firstly show that ginkgolide B promotes oligodendrocyte genesis and oligodendrocyte myelin following a WML, possibly involving the Akt and CREB pathways.


Assuntos
Diferenciação Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Ginkgolídeos/farmacologia , Lactonas/farmacologia , Células Precursoras de Oligodendrócitos/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transdução de Sinais , Substância Branca/patologia , Animais , Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Corpo Caloso/efeitos dos fármacos , Corpo Caloso/patologia , Memória/efeitos dos fármacos , Teste do Labirinto Aquático de Morris , Proteína Básica da Mielina/metabolismo , Bainha de Mielina/patologia , Células Precursoras de Oligodendrócitos/efeitos dos fármacos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Vacúolos/efeitos dos fármacos , Vacúolos/metabolismo , Substância Branca/efeitos dos fármacos , Substância Branca/fisiopatologia
10.
Brain ; 143(11): 3294-3307, 2020 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-33278823

RESUMO

For decades, it has been known that gliomas follow a non-random spatial distribution, appearing more often in some brain regions (e.g. the insula) compared to others (e.g. the occipital lobe). A better understanding of the localization patterns of gliomas could provide clues to the origins of these types of tumours, and consequently inform treatment targets. Following hypotheses derived from prior research into neuropsychiatric disease and cancer, gliomas may be expected to localize to brain regions characterized by functional hubness, stem-like cells, and transcription of genetic drivers of gliomagenesis. We combined neuroimaging data from 335 adult patients with high- and low-grade glioma to form a replicable tumour frequency map. Using this map, we demonstrated that glioma frequency is elevated in association cortex and correlated with multiple graph-theoretical metrics of high functional connectedness. Brain regions populated with putative cells of origin for glioma, neural stem cells and oligodendrocyte precursor cells, exhibited a high glioma frequency. Leveraging a human brain atlas of post-mortem gene expression, we found that gliomas were localized to brain regions enriched with expression of genes associated with chromatin organization and synaptic signalling. A set of glioma proto-oncogenes was enriched among the transcriptomic correlates of glioma distribution. Finally, a regression model incorporating connectomic, cellular, and genetic factors explained 58% of the variance in glioma frequency. These results add to previous literature reporting the vulnerability of hub regions to neurological disease, as well as provide support for cancer stem cell theories of glioma. Our findings illustrate how factors of diverse scale, from genetic to connectomic, can independently influence the anatomic localization of brain dysfunction.


Assuntos
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Encéfalo/patologia , Conectoma , Glioma/genética , Glioma/patologia , Algoritmos , Atlas como Assunto , Mapeamento Encefálico , Neoplasias Encefálicas/epidemiologia , Córtex Cerebral/patologia , Cromatina/genética , Regulação Neoplásica da Expressão Gênica/genética , Glioma/epidemiologia , Humanos , Células-Tronco Neoplásicas/patologia , Células-Tronco Neurais/patologia , Neuroimagem , Células Precursoras de Oligodendrócitos/patologia , Mudanças Depois da Morte , Sinapses/patologia
11.
Acta Neuropathol Commun ; 8(1): 120, 2020 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-32727582

RESUMO

Multiple system atrophy (MSA) is pathologically characterized by the presence of fibrillar α-synuclein-immunoreactive inclusions in oligodendrocytes. Although the myelinating process of oligodendrocytes can be observed in adult human brains, little is known regarding the presence of α-synuclein pathology in immature oligodendrocytes and how their maturation and myelination are affected in MSA brains. Recently, breast carcinoma amplified sequence 1 (BCAS1) has been found to be specifically expressed in immature oligodendrocytes undergoing maturation and myelination. Here, we analyzed the altered dynamics of oligodendroglial maturation in both MSA brains and primary oligodendroglial cell cultures which were incubated with α-synuclein pre-formed fibrils. The numbers of BCAS1-expressing oligodendrocytes that displayed a matured morphology negatively correlated with the density of pathological inclusions in MSA brains but not with that in Parkinson's disease and diffuse Lewy body disease. In addition, a portion of the BCAS1-expressing oligodendrocyte population showed cytoplasmic inclusions, which were labeled with antibodies against phosphorylated α-synuclein and cleaved caspase-9. Further in vitro examination indicated that the α-synuclein pre-formed fibrils induced cytoplasmic inclusions in the majority of BCAS1-expressing oligodendrocytes. In contrast, the majority of BCAS1-non-expressing mature oligodendrocytes did not develop inclusions on day 4 after maturation induction. Furthermore, exposure of α-synuclein pre-formed fibrils in the BCAS1-positive phase caused a reduction in oligodendroglial cell viability. Our results indicated that oligodendroglial maturation and myelination are impaired in the BCAS1-positive phase of MSA brains, which may lead to the insufficient replacement of defective oligodendrocytes. In vitro, the high susceptibility of BCAS1-expressing primary oligodendrocytes to the extracellular α-synuclein pre-formed fibrils suggests the involvement of insufficient oligodendroglial maturation in MSA disease progression and support the hypothesis that the BCAS1-positive oligodendrocyte lineage cells are prone to take up aggregated α-synuclein in vivo.


Assuntos
Atrofia de Múltiplos Sistemas/patologia , Células Precursoras de Oligodendrócitos/efeitos dos fármacos , Células Precursoras de Oligodendrócitos/patologia , alfa-Sinucleína/metabolismo , alfa-Sinucleína/toxicidade , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Humanos , Proteínas de Neoplasias , Proteínas do Tecido Nervoso , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/patologia , Ratos , Ratos Sprague-Dawley
12.
Adv Exp Med Biol ; 1234: 107-122, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32040858

RESUMO

Glioblastoma (GBM) develops from adult brain white matter and is the most common and lethal primary brain tumor, characterized by rapid growth and invasion. GBM tumors frequently spread into the contralateral hemisphere, including in the beginning of tumor development. However, after complete resection of the tumor mass and chemo-radiotherapy, GBM commonly recurs around the tumor removal site, suggesting that the microenvironment at the tumor border provides therapeutic resistance to GBM cells. To improve patient prognosis, understanding the microenvironment at the tumor border is critical. Several microRNAs (miRNAs) show higher expression at the tumor border, with the top three involved in oligodendrocyte differentiation. Oligodendrocyte progenitor cells (OPCs) may induce stemness and chemo-radioresistance in GBM cells, providing a supportive function to promote GBM. This review describes important features of OPCs and insights into the "border niche," a unique microenvironment that allows GBM cells to survive and recur at the tumor border.


Assuntos
Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Células Precursoras de Oligodendrócitos/patologia , Microambiente Tumoral , Humanos
13.
BMC Neurosci ; 20(1): 45, 2019 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-31470812

RESUMO

BACKGROUND: Systemic inflammation is an important risk factor for neurodevelopmental impairments in preterm infants. Premyelinating oligodendrocytes are main building blocks of white matter in preterm infants and vulnerable to oxidative stress and excitotoxic stress. Tumour necrosis factor-α (TNF-α) plays important roles in systemic inflammation and local inflammation leading to apoptosis of premyelinating oligodendrocytes and white matter injury (WMI) in brain tissue. This study was conducted to investigate whether etanercept, a TNF-α antagonist, could attenuate systemic lipopolysaccharide (LPS)-induced WMI in the immature brain. RESULTS: We found that intraperitoneal LPS administration caused systemic and local inflammation in brain tissue. Subsequent etanercept treatment significantly attenuated LPS-induced inflammation in brain tissue as well as in systemic circulation. Intraperitoneal LPS also induced microgliosis and astrocytosis in the cingulum and etanercept treatment reduced LPS-induced microgliosis and astrocytosis. Additionally, systemic LPS-induced apoptosis of oligodendrocyte precursor cells was observed, which was lessened by etanercept treatment. The concentration of etanercept in the CSF was higher when it was administrated with LPS than when administrated with a vehicle. CONCLUSIONS: It appears that etanercept reduce WMI in the neonatal rat brain via attenuation of systemic and local inflammation. This study provides preclinical data suggesting etanercept-mediated modulation of inflammation as a promising approach to reduce WMI caused by sepsis or necrotizing enterocolitis in preterm infants.


Assuntos
Encéfalo/patologia , Etanercepte/farmacologia , Gliose/prevenção & controle , Inflamação/prevenção & controle , Lipopolissacarídeos , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Substância Branca/patologia , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Etanercepte/líquido cefalorraquidiano , Feminino , Masculino , Células Precursoras de Oligodendrócitos/patologia , Ratos
14.
Acta Neuropathol ; 138(6): 987-1012, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31363836

RESUMO

Microglia are highly plastic immune cells which exist in a continuum of activation states. By shaping the function of oligodendrocyte precursor cells (OPCs), the brain cells which differentiate to myelin-forming cells, microglia participate in both myelin injury and remyelination during multiple sclerosis. However, the mode(s) of action of microglia in supporting or inhibiting myelin repair is still largely unclear. Here, we analysed the effects of extracellular vesicles (EVs) produced in vitro by either pro-inflammatory or pro-regenerative microglia on OPCs at demyelinated lesions caused by lysolecithin injection in the mouse corpus callosum. Immunolabelling for myelin proteins and electron microscopy showed that EVs released by pro-inflammatory microglia blocked remyelination, whereas EVs produced by microglia co-cultured with immunosuppressive mesenchymal stem cells promoted OPC recruitment and myelin repair. The molecular mechanisms responsible for the harmful and beneficial EV actions were dissected in primary OPC cultures. By exposing OPCs, cultured either alone or with astrocytes, to inflammatory EVs, we observed a blockade of OPC maturation only in the presence of astrocytes, implicating these cells in remyelination failure. Biochemical fractionation revealed that astrocytes may be converted into harmful cells by the inflammatory EV cargo, as indicated by immunohistochemical and qPCR analyses, whereas surface lipid components of EVs promote OPC migration and/or differentiation, linking EV lipids to myelin repair. Although the mechanisms through which the lipid species enhance OPC maturation still remain to be fully defined, we provide the first demonstration that vesicular sphingosine 1 phosphate stimulates OPC migration, the first fundamental step in myelin repair. From this study, microglial EVs emerge as multimodal and multitarget signalling mediators able to influence both OPCs and astrocytes around myelin lesions, which may be exploited to develop novel approaches for myelin repair not only in multiple sclerosis, but also in neurological and neuropsychiatric diseases characterized by demyelination.


Assuntos
Astrócitos/fisiologia , Doenças Desmielinizantes/fisiopatologia , Vesículas Extracelulares/fisiologia , Microglia/fisiologia , Bainha de Mielina/fisiologia , Remielinização/fisiologia , Animais , Astrócitos/patologia , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Técnicas de Cocultura , Corpo Caloso/patologia , Corpo Caloso/fisiopatologia , Doenças Desmielinizantes/patologia , Modelos Animais de Doenças , Vesículas Extracelulares/patologia , Inflamação/patologia , Inflamação/fisiopatologia , Lisofosfatidilcolinas , Masculino , Células-Tronco Mesenquimais/fisiologia , Camundongos Endogâmicos C57BL , Microglia/patologia , Bainha de Mielina/patologia , Neuroproteção/fisiologia , Células Precursoras de Oligodendrócitos/patologia , Células Precursoras de Oligodendrócitos/fisiologia , Ratos Sprague-Dawley
15.
Front Med ; 13(5): 531-539, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31313083

RESUMO

Glioblastoma (GBM) is the most common and lethal primary neoplasm in the central nervous system. Despite intensive treatment, the prognosis for patients with GBM remains poor, with a median survival of 14-16 months. 90% of GBMs are primary GBMs that are full-blown at diagnosis without evidences of a pre-existing less-malignant precursor lesion. Therefore, identification of the cell(s) of origin for GBM-the normal cell or cell type that acquires the initial GBM-promoting genetic hit(s)-is the key to the understanding of the disease etiology and the development of novel therapies. Neural stem cells and oligodendrocyte precursor cells are the two major candidates for the cell(s) of origin for GBM. Latest data from human samples have reignited the longstanding debate over which cells are the clinically more relevant origin for GBMs. By critically analyzing evidences for or against the candidacy of each cell type, we highlight the most recent progress and debate in the field, explore the clinical implications, and propose future directions toward early diagnosis and preventive treatment of GBMs.


Assuntos
Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Humanos , Células-Tronco Neurais/patologia , Células Precursoras de Oligodendrócitos/patologia
16.
Glia ; 67(9): 1775-1792, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31184779

RESUMO

Oligodendrocytes (OLs) provide the myelin sheath surrounding axons that propagates action potentials in the central nervous system (CNS). The metabolism of myelinated membranes and proteins is strictly regulated in the OLs and is closely associated with OL differentiation and maturation. The ubiquitination-associated proteasome and endosomal system have not yet been well studied during OL differentiation and maturation. Here, we determined the functions of the Lys63-linked ubiquitination (K63Ub) and K63-specific deubiquitination (DUB) systems regulated by BRCA1/BRCA2-containing complex subunit 3 (BRCC3) during OL differentiation. The competitive inhibition of K63Ub by overexpression of mutant ubiquitin (K63R) in oligodendrocyte precursor cells (OPCs) indicated that the two major CNS myelin proteins, myelin basic protein (MBP) and proteolipid protein (PLP), were upregulated in OLs derived from K63R OPCs. In contrast, the knockdown of BRCC3 (BRCC3-KD) through the application of lentivirus-mediated shRNA delivery system into OPCs suppressed OL differentiation by decreasing MBP expression and PLP production. Further immunoprecipitation assays revealed higher levels of sphingolipid GalC, MBP, and PLP, which were associated with K63Ub-immunoprecipitants and detected in endosome/lysosomal compartments, in BRCC3-KD OLs than those in OLs transfected with the scrambled shRNA (scramble OLs). The differentiation of OLs from BRCC3-KD OPCs was impaired in the demyelinating corpus callosum of rats receiving a cuprizone-containing diet. In the demyelinating tissues from human patients suffering from multiple sclerosis, we detected a decreased number of BRCC3-expressing OLs at the lesion site, accompanied by a greater number of OLs expressing EEA1 and K63Ub at high levels. Altogether, the counterbalance of the K63Ub machinery and BRCC3-triggered DUB machinery are important for the cellular trafficking of myelin proteins and OL differentiation.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Enzimas Desubiquitinantes/metabolismo , Neurogênese/fisiologia , Oligodendroglia/metabolismo , Ubiquitinação/fisiologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Células Cultivadas , Doenças Desmielinizantes/metabolismo , Doenças Desmielinizantes/patologia , Endossomos/metabolismo , Endossomos/patologia , Humanos , Lisossomos/metabolismo , Lisossomos/patologia , Masculino , Proteínas da Mielina/metabolismo , Células Precursoras de Oligodendrócitos/metabolismo , Células Precursoras de Oligodendrócitos/patologia , Oligodendroglia/patologia , Ratos Sprague-Dawley
17.
Folia Neuropathol ; 57(1): 24-35, 2019.
Artigo em Polonês | MEDLINE | ID: mdl-31038185

RESUMO

INTRODUCTION: This study was performed to determine whether the disturbed maturation of oligodendrocyte (OL) progenitors might be related to lipopolysaccharide (LPS)-induced hypomyelination. MATERIAL AND METHODS: We created organotypic cultures of forebrain slices from neonatal rats and explored the morphological changes of glial cells expressing tumour necrosis factor  (TNF-) following LPS exposure. RESULTS: We observed marked activation of glial fibrillary acidic protein-positive astrocytes and OX42-positive microglia co-labelled with TNF- four days following LPS exposure. Our results further demonstrated a reduced expression of O4-positive and O1-positive OL progenitors; moreover, we found that their morphologies were suggestive of degeneration (e.g., scanty, rounded bodies with short, fragmented processes and/or cytoplasmic condensation). At seven days following LPS exposure, astrocytes and microglia were still co-labelled for TNF-; however, the expression of O4-positive and O1-positive cells somewhat increased compared to the number observed at 4 days; despite remaining undifferentiated and exhibiting immature morphologies, the cells were likely indicative of regeneration. In contrast, O4-positive and O1-positive cells in controls were well-differentiated, displaying round, thick cell bodies and long, branching processes. CONCLUSIONS: In conclusion, maturation arrest and/or under-differentiation of OL progenitors commonly occur during regeneration: they may underlie the degeneration and consequent hypomyelination occurring late after injury, or apoptosis during the acute stage post-injury. Microglia and astrocytes expressing TNF- may also contribute to later myelination failure.


Assuntos
Inflamação/patologia , Fibras Nervosas Mielinizadas , Neurogênese/fisiologia , Células Precursoras de Oligodendrócitos/patologia , Animais , Animais Recém-Nascidos , Lipopolissacarídeos/toxicidade , Técnicas de Cultura de Órgãos , Prosencéfalo/embriologia , Ratos , Ratos Sprague-Dawley
18.
J Neuroimmunol ; 332: 176-186, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31075641

RESUMO

The myelin sheath is critical in maintaining normal functions of the adult central nervous system (CNS) and the loss of the myelin sheath results in various neurological diseases. Although remyelination is the intrinsic repair system against demyelination that new myelin sheath is formed around axons in the adult CNS, little has been reported on remyelination system in the medulla oblongata. In the present study, we showed that the proliferation of oligodendrocyte progenitor cells (OPCs) was increased in the medulla oblongata by lysophosphatidylcholine (LPC)-induced focal demyelination, but that of NSCs was not changed. The inhibition of vascular endothelial growth factor (VEGF)- and platelet-derived growth factor (PDGF)-signaling suppressed the proliferation of OPCs by LPC-induced demyelination. Thus, the present study indicates that resident OPCs contribute to focal remyelination and VEGF and PDGF signaling is required for the proliferation of OPCs in the medulla oblongata of the adult mouse.


Assuntos
Doenças Desmielinizantes/patologia , Bulbo/patologia , Células Precursoras de Oligodendrócitos/patologia , Fator de Crescimento Derivado de Plaquetas/fisiologia , Remielinização/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Animais , Divisão Celular/efeitos dos fármacos , Doenças Desmielinizantes/induzido quimicamente , Mesilato de Imatinib/farmacologia , Ventrículos Laterais/patologia , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Bainha de Mielina/fisiologia , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/patologia , Quinazolinas/farmacologia , Receptores do Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Organismos Livres de Patógenos Específicos
19.
Biomed Res Int ; 2019: 9524561, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31139661

RESUMO

The incidence of spinal cord injury (SCI) continues to increase; however, the involved mechanisms remain unclear. Anaphase promoting complex (APC) and its regulatory subunit Cdh1 play important roles in the growth, development, and repair of the central nervous system (CNS). Cdh1 is involved in the pathophysiological processes of neuronal apoptosis and astrocyte-reactive proliferation after ischemic brain injury, whereas the role played by APC-Cdh1 in the proliferation and activation of oligodendrocyte precursor cells (OPCs) after SCI remains unresolved. Using primary cultures of spinal oligodendrocyte precursor cells, we successfully established an in vitro mechanical stretch injury model to simulate SCI. Cell viability and proliferation were determined by MTT assay and flow cytometric analysis of the cell cycle. Real-time fluorescent quantitative PCR and Western blot analysis determined the mRNA and protein expression levels of Cdh1 and its downstream substrates Skp2 and Id2. Mechanical stretch injury decreased the proliferative activity of OPCs and enhanced cellular Cdh1 expression. Dampened expression of Cdh1 in primary OPCs significantly promoted proliferation and activation of OPCs after SCI. In addition, the expression of the downstream substrates of Cdh1, Skp2, and Id2 was decreased following mechanical injury, whereas adenovirus-mediated Cdh1 RNA interference increased postinjury expression of Skp2 and Id2. These findings suggest that APC-Cdh1 might be involved in regulating the proliferation and activation of OPCs after mechanical SCI. Moreover, degraded ubiquitination of the downstream substrates Skp2 and Id2 might play an important role, at least in part, in the beneficial effects of OPCs activity following SCI.


Assuntos
Ciclossomo-Complexo Promotor de Anáfase/metabolismo , Proteínas Cdh1/metabolismo , Células Precursoras de Oligodendrócitos/metabolismo , Células Precursoras de Oligodendrócitos/patologia , Estresse Mecânico , Adenoviridae/metabolismo , Animais , Proliferação de Células , Vetores Genéticos/metabolismo , Proteína 2 Inibidora de Diferenciação/metabolismo , Ratos Sprague-Dawley , Proteínas Quinases Associadas a Fase S/metabolismo , Especificidade por Substrato
20.
Brain Tumor Pathol ; 36(2): 63-73, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30968276

RESUMO

Glioblastoma (GBM) is a major malignant brain tumor developing in adult brain white matter, characterized by rapid growth and invasion. GBM cells spread into the contralateral hemisphere, even during early tumor development. However, after complete resection of tumor mass, GBM commonly recurs around the tumor removal cavity, suggesting that a microenvironment at the tumor border provides chemo-radioresistance to GBM cells. Thus, clarification of the tumor border microenvironment is critical for improving prognosis in GBM patients. MicroRNA (miRNA) expression in samples from the tumor, tumor border, and peripheral region far from tumor mass was compared, and five miRNAs showing characteristically higher expression in the tumor border were identified, with the top three related to oligodendrocyte differentiation. Pathologically, oligodendrocyte lineage cells increased in the border, but were rare in tumors. Macrophages/microglia also colocalized in the border area. Medium cultured with oligodendrocyte progenitor cells (OPCs) and macrophages induced stemness and chemo-radioresistance in GBM cells, suggesting that OPCs and macrophages/microglia constitute a special microenvironment for GBM cells at the tumor border. The supportive function of OPCs for GBM cells has not been discussed previously. OPCs are indispensable for GBM cells to establish special niches for chemo-radioresistance outside the tumor mass.


Assuntos
Glioblastoma/metabolismo , Glioblastoma/patologia , Células Precursoras de Oligodendrócitos/patologia , Neoplasias Encefálicas/patologia , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Linhagem da Célula , Humanos , Macrófagos/metabolismo , MicroRNAs/metabolismo , Microglia/patologia , Microglia/fisiologia , Recidiva Local de Neoplasia/patologia , Células-Tronco Neoplásicas/patologia , Células Precursoras de Oligodendrócitos/fisiologia , Oligodendroglia/patologia , Oligodendroglia/fisiologia , Microambiente Tumoral/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA