oligodendrocyte differentiation

少突胶质细胞分化
  • 文章类型: Journal Article
    少突胶质细胞(OLs)在中枢神经系统(CNS)中从少突胶质细胞前体细胞(OPCs)分化而来。脱髓鞘是许多神经系统疾病如多发性硬化(MS)和脑白质营养不良的共同特征。尽管在髓鞘损伤后可发生自发性髓鞘再生,然而,它通常是不足的,并可能导致加重的神经变性和神经残疾。我们先前的研究已经发现MEK/ERK途径在小鼠模型中负调节OPC到OL的分化和髓鞘再生。为了促进可能的临床评估,在这里,我们研究了几种已被FDA批准用于小鼠和人OPC-OL分化系统中癌症治疗的MEK抑制剂。曲美替尼,第一个FDA批准的MEK抑制剂,在所检查的四种MEK抑制剂中,在体外刺激OL产生方面表现出最佳效果。曲美替尼还显著增强MOG诱导的EAE模型和LPC诱导的局灶性脱髓鞘模型中的髓鞘再生。更令人兴奋,曲美替尼促进从人类胚胎干细胞(ESC)衍生的OPCs产生MBP+OL。机制研究表明,曲美替尼通过减少E2F1核易位和随后的转录活性来促进OL的产生。总之,我们的研究表明MEK/ERK在人和小鼠OL产生中具有相似的抑制作用。靶向MEK/ERK途径可能有助于开发新的疗法或重新利用现有药物治疗脱髓鞘疾病。
    Oligodendrocytes (OLs) are differentiated from oligodendrocyte precursor cells (OPCs) in the central nervous system (CNS). Demyelination is a common feature of many neurological diseases such as multiple sclerosis (MS) and leukodystrophies. Although spontaneous remyelination can happen after myelin injury, nevertheless, it is often insufficient and may lead to aggravated neurodegeneration and neurological disabilities. Our previous study has discovered that MEK/ERK pathway negatively regulates OPC-to-OL differentiation and remyelination in mouse models. To facilitate possible clinical evaluation, here we investigate several MEK inhibitors which have been approved by FDA for cancer therapies in both mouse and human OPC-to-OL differentiation systems. Trametinib, the first FDA approved MEK inhibitor, displays the best effect in stimulating OL generation in vitro among the four MEK inhibitors examined. Trametinib also significantly enhances remyelination in both MOG-induced EAE model and LPC-induced focal demyelination model. More exciting, trametinib facilitates the generation of MBP+ OLs from human embryonic stem cells (ESCs)-derived OPCs. Mechanism study indicates that trametinib promotes OL generation by reducing E2F1 nuclear translocation and subsequent transcriptional activity. In summary, our studies indicate a similar inhibitory role of MEK/ERK in human and mouse OL generation. Targeting the MEK/ERK pathway might help to develop new therapies or repurpose existing drugs for demyelinating diseases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    神经元信号如何影响脑髓鞘形成仍然知之甚少。我们显示神经元RHEB-mTORC1-DLK1轴失调损害脑髓鞘形成。神经元RhebcKO损害少突胶质细胞分化/髓鞘形成,具有激活的神经元表达的印迹基因Dlk1。神经元Dlk1cKO改善神经元RhebcKO小鼠的髓鞘形成缺陷,表明激活的神经元Dlk1表达有助于由RhebcKO引起的髓鞘形成受损。RhebcKO对Dlk1表达的影响由mTORC1介导;神经元mTorcKO和RaptorcKO以及对mTORC1的药理学抑制概括了神经元Dlk1表达的升高。我们证明了DLK1的分泌形式和膜结合的DLK1均抑制培养的少突胶质细胞前体细胞分化为表达髓磷脂蛋白的少突胶质细胞。最后,转基因小鼠中Dlk1的神经元表达减少了成熟少突胶质细胞的形成和髓鞘形成。这项研究确定了Dlk1是少突胶质细胞髓鞘形成的抑制剂,以及将神经元信号传导改变与少突胶质细胞功能障碍联系起来的机制。
    How neuronal signaling affects brain myelination remains poorly understood. We show dysregulated neuronal RHEB-mTORC1-DLK1 axis impairs brain myelination. Neuronal Rheb cKO impairs oligodendrocyte differentiation/myelination, with activated neuronal expression of the imprinted gene Dlk1. Neuronal Dlk1 cKO ameliorates myelination deficit in neuronal Rheb cKO mice, indicating that activated neuronal Dlk1 expression contributes to impaired myelination caused by Rheb cKO. The effect of Rheb cKO on Dlk1 expression is mediated by mTORC1; neuronal mTor cKO and Raptor cKO and pharmacological inhibition of mTORC1 recapitulate elevated neuronal Dlk1 expression. We demonstrate that both a secreted form of DLK1 and a membrane-bound DLK1 inhibit the differentiation of cultured oligodendrocyte precursor cells into oligodendrocytes expressing myelin proteins. Finally, neuronal expression of Dlk1 in transgenic mice reduces the formation of mature oligodendrocytes and myelination. This study identifies Dlk1 as an inhibitor of oligodendrocyte myelination and a mechanism linking altered neuronal signaling with oligodendrocyte dysfunction.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    少突胶质细胞(OL)死亡和髓鞘再生失败导致多发性硬化(MS)及其动物模型的进行性神经功能缺损,实验性自身免疫性脑脊髓炎(EAE)。苦参碱(MAT),一种来自苦参根的喹诺酮苷生物碱成分,具有有效抑制中枢神经系统(CNS)炎症和促进神经再生的能力。在本研究中,我们探讨了其对Wnt/β-catenin/TCF7L2通路的调控机制,髓鞘形成的负调节剂,在MOG35--55肽诱导的EAE中。我们的结果清楚地表明,MAT治疗降低了EAE小鼠CNS中Wnt3a和β-catenin的激活,伴随着Wnt3a/β-catenin途径的两个靶基因GSK3β的激活和cyclinD1和Axin2的表达降低。此外,MAT增加了OL成熟和髓鞘形成,如NG2+Olig2+细胞的数量减少和MBP+和CC1+Olig2+细胞的数量增加所证明的。一起来看,这些发现表明MAT治疗促进了OLs的成熟和髓鞘修复,与Wnt/β-catenin/TCF7L2信号通路的调控密切相关。
    Oligodendrocyte (OL) death and remyelination failure lead to progressive neurological deficits in multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE). Matrine (MAT), a quinolizidine alkaloid component derived from the root of Sophora flavescens, has the capacity to effectively inhibit central nervous system (CNS) inflammation and to promote neuroregeneration. In the present study we explored its regulatory mechanism on the Wnt/β-catenin/TCF7L2 pathway, a negative modulator for myelination, in MOG35--55 peptide-induced EAE. Our results clearly indicate that MAT treatment reduced the activation of Wnt3a and β-catenin in the CNS of EAE mice, accompanied by the activation of GSK3β and decreased expression of cyclin D1 and Axin2, two target genes of the Wnt3a/β-catenin pathway. In addition, MAT increased OL maturation and myelination, as evidenced by the decreased number of NG2+Olig2+ cells and the increased numbers of MBP+ and CC1+Olig2+ cells. Taken together, these findings indicate that MAT treatment promoted the maturation of OLs and myelin repair, which is closely related to the modulation of the Wnt/β-catenin/TCF7L2 signaling pathway.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    已知富含亮氨酸的神经胶质瘤灭活蛋白1(LGI1)在常染色体显性遗传颞叶外侧癫痫(ADLTE)中起关键作用。ADLTE是一种遗传性疾病,其特征是局灶性癫痫发作,具有独特的听觉或失语症状。已经报道了Lgi1基因上的大量突变,并且被认为是ADLTE的遗传原因。我们发现了一个新的错义突变,c.152A>G(p。Asp51Gly),来自中国ADLTE患者的Lgi1,该患者表现出运动失衡和白质减少。然而,目前尚不清楚突变LGI1如何导致分子和细胞水平的白质异常.这里,我们产生了一个带有Lgi1突变的敲入小鼠。我们发现Lgi1D51G/D51G小鼠表现出受损的白质和运动协调缺陷。我们观察到Lgi1D51G/D51G小鼠在白质中显示出数量减少的成熟少突胶质细胞(OL)和缺乏OL分化。然而,Lgi1D51G/D51G小鼠的少突胶质细胞前体细胞群没有受到影响。机械上,我们发现Lgi1D51G突变导致mTOR信号传导改变,并导致Sox10水平降低.鉴于Sox10是控制OL分化的关键转录因子,我们的结果强烈提示Lgi1D51G突变可能通过抑制Sox10依赖性的OL分化和中枢神经系统髓鞘形成而导致白质异常.
    Leucine-rich glioma-inactivated protein 1 (LGI1) is known to play a key role in autosomal dominant lateral temporal lobe epilepsy (ADLTE). The ADLTE is an inherited disease characterized by focal seizures with distinctive auditory or aphasic symptoms. A large number of mutations on the Lgi1 gene have been reported and are believed to be the genetic cause for ADLTE. We identified a novel missense mutation, c.152A>G (p.Asp51Gly), on Lgi1 from a Chinese ADLTE patient who manifests locomotor imbalance and white matter reduction. However, it remains unknown how mutant LGI1 causes white matter abnormalities at molecular and cellular levels. Here, we generated a knock-in mouse bearing this Lgi1 mutation. We found that Lgi1D51G / D51G mice exhibited impaired defective white matter and motor coordination. We observed that Lgi1D51G / D51G mice displayed a reduced number of mature oligodendrocytes (OLs) and deficient OL differentiation in the white matter. However, the population of oligodendrocyte precursor cells was not affected in Lgi1D51G / D51G mice. Mechanistically, we showed that the Lgi1D51G mutation resulted in altered mTOR signaling and led to decreased levels of Sox10. Given that Sox10 is a key transcriptional factor to control OL differentiation, our results strongly suggest that the Lgi1D51G mutation may cause white matter abnormalities via inhibiting Sox10-dependent OL differentiation and myelination in the central nervous system.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    随着癌症治疗技术的提高,化疗引起的脑白质损伤引起的认知障碍受到越来越多的关注.已显示富马酸氯马斯汀可增强铜氮或缺氧诱导的脱髓鞘小鼠模型中的白质完整性。然而,氯马斯汀是否有利于逆转化疗诱导的认知障碍仍有待研究.在这项研究中,化疗后,小鼠口服氯马斯汀。采用开场试验和Morris水迷宫试验评价其焦虑,运动活动和认知功能。LuxolFastBlue染色和透射电子显微镜用于检测髓鞘的形态损伤。通过免疫荧光和蛋白质印迹观察到成熟少突胶质细胞和轴突的脱髓鞘和损伤。在化疗的小鼠中,氯马斯汀显着改善了其认知功能并改善了白质损伤。氯马斯汀增强髓鞘形成,促进少突胶质细胞前体细胞分化,并增加call体和海马中神经丝200蛋白的水平。我们得出的结论是,氯马斯汀通过改善脑白质完整性来挽救化疗引起的认知功能损害。髓鞘再生,氯马斯汀促进少突胶质细胞分化和神经丝蛋白的增加是逆转化疗引起的认知功能障碍的潜在策略。
    With the improvement of cancer treatment techniques, increasing attention has been given to chemotherapy-induced cognitive impairment through white matter injury. Clemastine fumarate has been shown to enhance white matter integrity in cuprizone- or hypoxia-induced demyelination mouse models. However, whether clemastine can be beneficial for reversing chemotherapy-induced cognitive impairment remains unexplored. In this study, the mice received oral administration of clemastine after chemotherapy. The open-field test and Morris water maze test were used to evaluate their anxiety, locomotor activity and cognitive function. Luxol Fast Blue staining and transmission electron microscopy were used to detect the morphological damage to the myelin. Demyelination and damage to the mature oligodendrocytes and axons were observed by immunofluorescence and western blotting. Clemastine significantly improved their cognitive function and ameliorated white matter injury in the chemotherapy-treated mice. Clemastine enhanced myelination, promoted oligodendrocyte precursor cell differentiation and increased the neurofilament 200 protein levels in the corpus callosum and hippocampus. We concluded that clemastine rescues cognitive function damage caused by chemotherapy through improving white matter integrity. Remyelination, oligodendrocyte differentiation and the increase of neurofilament protein promoted by clemastine are potential strategies for reversing the cognitive dysfunction caused by chemotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    Sox10 is a well known factor to control oligodendrocyte (OL) differentiation, and its expression is regulated by Olig2. As an important protein kinase, Akt has been implicated in diseases with white matter abnormalities. To study whether and how Akt may regulate OL development, we generated OL lineage cell-specific Akt1/Akt2/Akt3 triple conditional knock-out (Akt cTKO) mice. Both male and female mice were used. These mutants exhibit a complete loss of mature OLs and unchanged apoptotic cell death in the CNS. We show that the deletion of Akt three isoforms causes downregulation of Sox10 and decreased levels of phosphorylated FoxO1 in the brain. In vitro analysis reveals that the expression of FoxO1 with mutations on phosphorylation sites for Akt significantly represses the Sox10 promoter activity, suggesting that phosphorylation of FoxO1 by Akt is important for Sox10 expression. We further demonstrate that mutant FoxO1 without Akt phosphorylation epitopes is enriched in the Sox10 promoter. Together, this study identifies a novel FoxO1 phosphorylation-dependent mechanism for Sox10 expression and OL differentiation.SIGNIFICANCE STATEMENT Dysfunction of Akt is associated with white matter diseases including the agenesis of the corpus callosum. However, it remains unknown whether Akt plays an important role in oligodendrocyte differentiation. To address this question, we generated oligodendrocyte lineage cell-specific Akt1/Akt2/Akt3 triple-conditional knock-out mice. Akt mutants exhibit deficient white matter development, loss of mature oligodendrocytes, absence of myelination, and unchanged apoptotic cell death in the CNS. We demonstrate that deletion of Akt three isoforms leads to downregulation of Sox10, and that phosphorylation of FoxO1 by Akt is critical for Sox10 expression. Together, these findings reveal a novel mechanism to regulate Sox10 expression. This study may provide insights into molecular mechanisms for neurodevelopmental diseases caused by dysfunction of protein kinases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    γ-分泌酶亚基的突变与神经系统疾病相关。而γ-分泌酶在神经发生中的作用已被深入研究,对其在星形胶质细胞生成中的作用知之甚少。最近的证据表明,星形胶质细胞可以从少突胶质细胞前体细胞(OPC)产生。然而,目前尚不清楚是什么机制可以控制OPCs分化为星形胶质细胞。为了解决上述问题,我们产生了两个独立的少突胶质细胞谱系特异性早老素增强子2(Pen-2)条件性KO小鼠系。使用雄性和雌性小鼠。在这里,我们证明了由Olig1-Cre或NG2-CreERT2介导的Pen-2的条件性失活导致星形胶质细胞的生成增强。谱系追踪实验表明,在Pen-2条件性KO小鼠中,异常产生的星形胶质细胞源自CNS中表达Cre的OPC。机制分析显示Pen-2的缺失抑制Notch信号传导以上调信号转导子和转录激活因子3,这触发GFAP的激活以促进星形胶质细胞分化。一起,这些新发现表明,Pen-2通过信号转导和转录激活因子3信号调节OPCs星形胶质细胞的规格。意义陈述星形胶质细胞和少突胶质细胞(OLs)在脑中起关键作用。最近的证据表明,星形胶质细胞可以从OL前体细胞(OPC)中产生。然而,目前尚不清楚OPCs分化为星形胶质细胞的机制是什么.在这项研究中,我们利用了OL谱系细胞特异性早老素增强子2(Pen-2)条件KO小鼠。我们表明,Pen-2的缺失导致CNS中OPCs的星形胶质细胞分化显着增强。机制分析显示Pen-2的缺失抑制Hes1并激活信号转导和转录激活因子3以触发促进星形胶质细胞分化的GFAP激活。总的来说,这项研究从OPCs中鉴定了Pen-2在星形胶质细胞生成中的新功能。
    Mutations on γ-secretase subunits are associated with neurologic diseases. Whereas the role of γ-secretase in neurogenesis has been intensively studied, little is known about its role in astrogliogenesis. Recent evidence has demonstrated that astrocytes can be generated from oligodendrocyte precursor cells (OPCs). However, it is not well understood what mechanism may control OPCs to differentiate into astrocytes. To address the above questions, we generated two independent lines of oligodendrocyte lineage-specific presenilin enhancer 2 (Pen-2) conditional KO mice. Both male and female mice were used. Here we demonstrate that conditional inactivation of Pen-2 mediated by Olig1-Cre or NG2-CreERT2 causes enhanced generation of astrocytes. Lineage-tracing experiments indicate that abnormally generated astrocytes are derived from Cre-expressing OPCs in the CNS in Pen-2 conditional KO mice. Mechanistic analysis reveals that deletion of Pen-2 inhibits the Notch signaling to upregulate signal transducer and activator of transcription 3, which triggers activation of GFAP to promote astrocyte differentiation. Together, these novel findings indicate that Pen-2 regulates the specification of astrocytes from OPCs through the signal transducer and activator of transcription 3 signaling.SIGNIFICANCE STATEMENT Astrocytes and oligodendrocyte (OLs) play critical roles in the brain. Recent evidence has demonstrated that astrocytes can be generated from OL precursor cells (OPCs). However, it remains poorly understood what mechanism governs the differentiation of OPCs into astrocytes. In this study, we took advantage of OL lineage cells specific presenilin enhancer 2 (Pen-2) conditional KO mice. We show that deletion of Pen-2 leads to dramatically enhanced astrocyte differentiation from OPCs in the CNS. Mechanistic analysis reveals that deletion of Pen-2 inhibits Hes1 and activates signal transducer and activator of transcription 3 to trigger GFAP activation which promotes astrocyte differentiation. Overall, this study identifies a novel function of Pen-2 in astrogliogenesis from OPCs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    Promoting oligodendrocyte (OL) differentiation represents a promising option for remyelination therapy for treating the demyelinating disease multiple sclerosis (MS). The Wnt effector transcription factor 7-like 2 (TCF7l2) was upregulated in MS lesions and had been proposed to inhibit OL differentiation. Recent data suggest the opposite yet underlying mechanisms remain elusive. Here, we unravel a previously unappreciated function of TCF7l2 in controlling autocrine bone morphogenetic protein (BMP)4-mediated signaling. Disrupting TCF7l2 in mice of both sexes results in oligodendroglial-specific BMP4 upregulation and canonical BMP4 signaling activation in vivo Mechanistically, TCF7l2 binds to Bmp4 gene regulatory element and directly represses its transcriptional activity. Functionally, enforced TCF7l2 expression promotes OL differentiation by reducing autocrine BMP4 secretion and dampening BMP4 signaling. Importantly, compound genetic disruption demonstrates that oligodendroglial-specific BMP4 deletion rescues arrested OL differentiation elicited by TCF7l2 disruption in vivo Collectively, our study reveals a novel connection between TCF7l2 and BMP4 in oligodendroglial lineage and provides new insights into augmenting TCF7l2 for promoting remyelination in demyelinating disorders such as MS.SIGNIFICANCE STATEMENT Incomplete or failed myelin repairs, primarily resulting from the arrested differentiation of myelin-forming oligodendrocytes (OLs) from oligodendroglial progenitor cells, is one of the major reasons for neurologic progression in people affected by multiple sclerosis (MS). Using in vitro culture systems and in vivo animal models, this study unraveled a previously unrecognized autocrine regulation of bone morphogenetic protein (BMP)4-mediated signaling by the Wnt effector transcription factor 7-like 2 (TCF7l2). We showed for the first time that TCF7l2 promotes oligodendroglial differentiation by repressing BMP4-mediated activity, which is dysregulated in MS lesions. Our study suggests that elevating TCF7l2 expression may be possible in overcoming arrested oligodendroglial differentiation as observed in MS patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    In the developing spinal cord, the majority of oligodendrocyte progenitor cells (OPCs) are induced in the ventral neuroepithelium under the control of the Sonic Hedgehog (Shh) signaling pathway, whereas a small subset of OPCs are generated from the dorsal neuroepithelial cells independent of the Shh pathway. Although dorsally-derived OPCs (dOPCs) have been shown to participate in local axonal myelination in the dorsolateral regions during development, it is not known whether they are capable of migrating into the ventral region and myelinating ventral axons. In this study, we confirmed and extended the previous study on the developmental potential of dOPCs in the absence of ventrally-derived OPCs (vOPCs). In Nestin-Smo conditional knockout (cKO) mice, when ventral oligodendrogenesis was blocked, dOPCs were found to undergo rapid amplification, spread to ventral spinal tissue, and eventually differentiated into myelinating OLs in the ventral white matter with a temporal delay, providing genetic evidence that dOPCs are capable of myelinating ventral axons in the mouse spinal cord.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Focal inflammation and remyelination failure are major hallmarks of multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE). In this study, we found that leonurine, a bioactive alkaloid, alleviated EAE disease severity along with reduced central nervous system inflammation and myelin damage. During the pathogenesis of EAE, leonurine dramatically suppressed the recruitment of encephalitogenic T cells into the central nervous system, whereas did not impair periphery immune responses and microglia activation. Mechanistically, leonurine protected mice against demyelination along with enhanced remyelination through promoting the maturation of oligodendrocytes in both EAE and cuprizone-induced demyelination mouse models. Moreover, we identified that the expression of demethylase jumonji domain-containing protein D3 was significantly enhanced upon treatment of leonurine, which suppressed the trimethylation of histone H3 lysine-27 and enhanced oligodendrocyte maturation accordingly. Collectively, our study identified the therapeutic effect of leonurine on EAE model, which potentially represents a promising therapeutic strategy for multiple sclerosis, even other demyelination disorders.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号