remyelination

髓鞘再生
  • 文章类型: Journal Article
    周围神经损伤(PNI)是一个重大的临床挑战,特别是在轴突髓鞘再生和再生受损的老年人群。开发增强这些过程的疗法对于改善PNI修复结果至关重要。谷氨酸羧肽酶II(GCPII)是一种神经肽酶,通过其酶切丰富的神经肽N-乙酰基天冬氨酰谷氨酸(NAAG)释放谷氨酸来调节谷氨酸信号传导中起关键作用。在PNS内,GCPII在施万细胞和活化的巨噬细胞中表达,其表达随着衰老而扩增。在这项研究中,我们探索了PNI后抑制GCPII活性的治疗潜力.我们报道了PNI后GCPII蛋白和活性的显著上调,其通过有效和选择性的GCPII抑制剂2-(膦酰基甲基)-戊二酸(2-PMPA)进行归一化。体外,2-PMPA强烈增强背根神经节(DRG)外植体的髓鞘形成。在体内,在老年小鼠中使用坐骨神经挤压损伤模型,2-PMPA加速髓鞘再生,髓鞘厚度增加和髓鞘再生轴突数量增加。这些发现表明,GCPII抑制可能是一种有希望的治疗策略,以增强髓鞘再生并可能改善PNI后的功能恢复。这在该过程受损的老年PNI患者中尤其重要。
    Peripheral nerve injuries (PNIs) represent a significant clinical challenge, particularly in elderly populations where axonal remyelination and regeneration are impaired. Developing therapies to enhance these processes is crucial for improving PNI repair outcomes. Glutamate carboxypeptidase II (GCPII) is a neuropeptidase that plays a pivotal role in modulating glutamate signaling through its enzymatic cleavage of the abundant neuropeptide N-acetyl aspartyl glutamate (NAAG) to liberate glutamate. Within the PNS, GCPII is expressed in Schwann cells and activated macrophages, and its expression is amplified with aging. In this study, we explored the therapeutic potential of inhibiting GCPII activity following PNI. We report significant GCPII protein and activity upregulation following PNI, which was normalized by the potent and selective GCPII inhibitor 2-(phosphonomethyl)-pentanedioic acid (2-PMPA). In vitro, 2-PMPA robustly enhanced myelination in dorsal root ganglion (DRG) explants. In vivo, using a sciatic nerve crush injury model in aged mice, 2-PMPA accelerated remyelination, as evidenced by increased myelin sheath thickness and higher numbers of remyelinated axons. These findings suggest that GCPII inhibition may be a promising therapeutic strategy to enhance remyelination and potentially improve functional recovery after PNI, which is especially relevant in elderly PNI patients where this process is compromised.
    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
    背景:多发性硬化症(MS)的治疗目标传统上被分为两种不同的途径:以免疫调节为中心的干预措施和促再生策略。多年来,人们一直认为少突胶质细胞祖细胞(OPCs)仅考虑其在中枢神经系统(CNS)中产生少突胶质细胞和髓磷脂的潜力。然而,积累的数据阐明了OPC的多方面作用,包括它们的免疫调节功能,将它们定位为中枢神经系统免疫景观的主要成分。
    方法:在这篇综述中,我们将讨论这两种治疗方法是如何融合的。我们提出了一个模型,通过该模型(1)慢性发炎的CNS中OPCs的适当的髓鞘形成免疫功能需要炎症,(2)OPCs的免疫功能对其分化和促进髓鞘再生的能力至关重要。该模型强调了OPCs前髓鞘形成和免疫调节功能之间的相互作用。此外,我们回顾了抗炎和促炎干预对OPCs的具体影响,表明免疫抑制对OPCs的分化和免疫功能产生不利影响。
    结论:我们建议采用多系统治疗方法,这不需要一维聚焦,而是需要OPCs前髓鞘形成和免疫调节功能之间的和谐平衡。
    BACKGROUND: Multiple sclerosis (MS) therapeutic goals have traditionally been dichotomized into two distinct avenues: immune-modulatory-centric interventions and pro-regenerative strategies. Oligodendrocyte progenitor cells (OPCs) were regarded for many years solely in concern to their potential to generate oligodendrocytes and myelin in the central nervous system (CNS). However, accumulating data elucidate the multifaceted roles of OPCs, including their immunomodulatory functions, positioning them as cardinal constituents of the CNS\'s immune landscape.
    METHODS: In this review, we will discuss how the two therapeutic approaches converge. We present a model by which (1) an inflammation is required for the appropriate pro-myelinating immune function of OPCs in the chronically inflamed CNS, and (2) the immune function of OPCs is crucial for their ability to differentiate and promote remyelination. This model highlights the reciprocal interactions between OPCs\' pro-myelinating and immune-modulating functions. Additionally, we review the specific effects of anti- and pro-inflammatory interventions on OPCs, suggesting that immunosuppression adversely affects OPCs\' differentiation and immune functions.
    CONCLUSIONS: We suggest a multi-systemic therapeutic approach, which necessitates not a unidimensional focus but a harmonious balance between OPCs\' pro-myelinating and immune-modulatory functions.
    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
    用于治疗脱髓鞘疾病如多发性硬化症的细胞疗法因供体少突胶质细胞细胞制剂的低存活率而受到阻碍。导致治疗效果有限。过度的细胞死亡导致细胞内同种抗原的释放,这可能会加剧局部炎症,并可能使移植物最终发生排斥反应。这里,我们设计了具有可调粘弹性和生物活性的创新的细胞指导性剪切稀化水凝胶(STHs),用于将原代人少突胶质细胞祖细胞(hOPCs)微创递送至颤抖/rag2小鼠的大脑,先天性骨髓增生异常疾病的模型。STHs能够固定促生存信号,包括重组设计的bidomain肽和血小板衍生的生长因子。值得注意的是,STHs显著降低hOPCs的死亡率,促进髓鞘少突胶质细胞的产生,植入后12周,小鼠大脑的髓鞘形成增强。我们的结果证明了负载有生物线索的STHs改善细胞疗法治疗破坏性脊髓病的潜力。
    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.
    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
    脱髓鞘的特征在于髓鞘的破坏和髓鞘形成的紊乱。目前,没有有效的治疗方法。小胶质细胞,尤其是抗炎表型小胶质细胞是髓鞘再生的关键.半乳糖凝集素-3(Gal-3),已知可以调节小胶质细胞的激活,与髓鞘形成有关。在这项研究中,我们旨在阐明Gal-3在髓鞘形成过程中的作用,并探讨rGal-3在髓鞘再生过程中的应用效率和作用机制.我们招募了Gal-3敲除(Lgals3KO)小鼠,并证明了Lgals3KO在自发性髓鞘形成过程中会引起脱髓鞘。我们进行了铜宗(CPZ)中毒模型,发现Lgals3KO加重了脱髓鞘病变,并有利于小胶质细胞促炎表型极化。重组Gal-3(rGal-3)施用减轻CPZ中毒并驱动小胶质细胞朝向抗炎表型。此外,RNA测序结果揭示了Gal-3与PPARγ-CD36轴之间的相关性。因此,我们进行了SSO和GW9662给药,以抑制PPARγ-CD36轴的激活,并发现rGal-3给药通过调节PPARγ-CD36轴来调节小胶质细胞表型极化.一起,我们的研究结果强调了Gal-3在髓鞘形成中的重要性,并为rGal-3通过PPARγ-CD36轴调节小胶质细胞抗炎表型极化提供了见解.
    Demyelination is characterized by disruption of myelin sheath and disorders in myelin formation. Currently, there are no effective therapeutic treatments available. Microglia, especially anti-inflammatory phenotype microglia are critical for remyelination. Galectin-3 (Gal-3), which is known to modulate microglia activation, is correlated with myelination. In this study, we aimed to elucidate the roles of Gal-3 during myelin formation and explore the efficiency and mechanism of rGal-3 administration in remyelination. We enrolled Gal-3 knockout (Lgals3 KO) mice and demonstrated Lgals3 KO causes demyelination during spontaneous myelinogenesis. We performed a cuprizone (CPZ) intoxication model and found Lgals3 KO aggravates demyelinated lesions and favors microglial pro-inflammatory phenotype polarization. Recombinant Gal-3 (rGal-3) administration alleviates CPZ intoxication and drives microglial towards anti-inflammatory phenotype. Additionally, RNA sequencing results reveal the correlation between Gal-3 and the PPARγ-CD36 axis. Thus, we performed SSO and GW9662 administration to inhibit the activation of the PPARγ-CD36 axis and found that rGal-3 administration modulates microglial phenotype polarization by regulating the PPARγ-CD36 axis. Together, our findings highlight the importance of Gal-3 in myelination and provide insights into rGal-3 administration for modulating microglial anti-inflammatory phenotype polarization through the PPARγ-CD36 axis.
    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
    治疗进行性多发性硬化症(MS)的策略仍然有限。这里,我们发现miR-145-5p在继发性进展型MS患者的慢性病变组织中含量过多.我们在miR-145敲除小鼠中诱导了急性和慢性脱髓鞘,以确定其对髓鞘再生失败的贡献。急性脱髓鞘后,未检测到miR-145丢失的优势.然而,慢性脱髓鞘后,miR-145丢失的动物表现出增加的髓鞘再生和功能恢复,与野生型动物相比,call体内星形胶质细胞和小胶质细胞的存在发生了变化。miR-145敲除动物中的这种改善的反应与miR-145-5p在具有慢性铜宗暴露的野生型动物中的病理上调相吻合。平行人类慢性病变。此外,miR-145在少突胶质细胞(OLs)中特异性过表达严重阻碍分化并负面影响存活。RNAseq分析显示这些细胞中的转录组改变,与髓鞘形成有关的主要途径下调。我们的数据表明,miR-145-5p的病理积累是慢性脱髓鞘的一个显著特征,并且与髓鞘再生的失败密切相关。可能是由于OL分化的抑制以及其他神经胶质细胞的改变。这反映在慢性MS病变中,因此,miR-145-5p在MS的进行性形式中充当潜在的相关治疗靶标。
    Strategies for treating progressive multiple sclerosis (MS) remain limited. Here, we found that miR-145-5p is overabundant uniquely in chronic lesion tissues from secondary progressive MS patients. We induced both acute and chronic demyelination in miR-145 knockout mice to determine its contributions to remyelination failure. Following acute demyelination, no advantage to miR-145 loss could be detected. However, after chronic demyelination, animals with miR-145 loss demonstrated increased remyelination and functional recovery, coincident with altered presence of astrocytes and microglia within the corpus callosum relative to wild-type animals. This improved response in miR-145 knockout animals coincided with a pathological upregulation of miR-145-5p in wild-type animals with chronic cuprizone exposure, paralleling human chronic lesions. Furthermore, miR-145 overexpression specifically in oligodendrocytes (OLs) severely stunted differentiation and negatively impacted survival. RNAseq analysis showed altered transcriptome in these cells with downregulated major pathways involved in myelination. Our data suggest that pathological accumulation of miR-145-5p is a distinctive feature of chronic demyelination and is strongly implicated in the failure of remyelination, possibly due to the inhibition of OL differentiation together with alterations in other glial cells. This is mirrored in chronic MS lesions, and thus miR-145-5p serves as a potential relevant therapeutic target in progressive forms of MS.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Letter
    皮质脊髓束神经元脱髓鞘有助于皮质卒中后的长期残疾。尽管如此,卒中后髓鞘丢失尚未作为治疗靶点,到目前为止。我们假设抗体介导的Nogo受体相互作用蛋白(LINGO-1,富含亮氨酸的重复序列和含有免疫球蛋白结构域的Nogo受体相互作用蛋白)的抑制可能会抵消髓鞘丢失,增强髓鞘再生和轴突生长,从而促进中风后的功能恢复。为了验证这个假设,对小鼠进行光血栓形成性中风,并接受抗LINGO-1抗体(n=19)或对照治疗(n=18).进行行为测试以评估抗LINGO-1治疗对功能恢复的影响。中风后七周,进行免疫组织化学分析以分析抗LINGO-1治疗对皮质脊髓束神经元髓鞘形成和轴突丢失的影响,少突胶质细胞的增殖和神经发生。抗LINGO-1治疗导致显著改善的功能恢复(p<0.0001,重复测量方差分析),同侧半球海马和脑室下区的神经发生增加(p=0.0094和p=0.032,t检验)。值得注意的是,我们观察到髓磷脂的显着增加(p=0.0295,t检验),抗LINGO-1治疗小鼠同侧内囊内的血小板衍生生长因子受体α阳性少突胶质细胞前体细胞(p=0.0356,t检验)和髓鞘腺瘤性结肠息肉阳性细胞(p=0.0021,t检验)。总之,我们确定抗LINGO-1是第一个对抗卒中后皮质脊髓束神经元脱髓鞘的神经再生治疗,可能是由于髓鞘前体细胞的增殖增加,从而提高功能恢复。最重要的是,我们的研究将髓磷脂丢失作为卒中后的一个新的治疗靶点.
    Demyelination of corticospinal tract neurons contributes to long-term disability after cortical stroke. Nonetheless, poststroke myelin loss has not been addressed as a therapeutic target, so far. We hypothesized that an antibody-mediated inhibition of the Nogo receptor-interacting protein (LINGO-1, leucine-rich repeat and immunoglobulin domain-containing Nogo receptor-interacting protein) may counteract myelin loss, enhance remyelination and axonal growth, and thus promote functional recovery following stroke. To verify this hypothesis, mice were subjected to photothrombotic stroke and received either an antibody against LINGO-1 (n = 19) or a control treatment (n = 18). Behavioral tests were performed to assess the effects of anti-LINGO-1 treatment on the functional recovery. Seven weeks after stroke, immunohistochemical analyses were performed to analyze the effect of anti-LINGO-1 treatment on myelination and axonal loss of corticospinal tract neurons, proliferation of oligodendrocytes and neurogenesis. Anti-LINGO-1 treatment resulted in significantly improved functional recovery (p < 0.0001, repeated measures analysis of variance), and increased neurogenesis in the hippocampus and subventricular zone of the ipsilateral hemisphere (p = 0.0094 and p = 0.032, t-test). Notably, we observed a significant increase in myelin (p = 0.0295, t-test), platelet-derived growth factor receptor α-positive oligodendrocyte precursor cells (p = 0.0356, t-test) and myelinating adenomatous polyposis coli-positive cells within the ipsilateral internal capsule of anti-LINGO-1-treated mice (p = 0.0021, t-test). In conclusion, we identified anti-LINGO-1 as the first neuroregenerative treatment that counteracts poststroke demyelination of corticospinal tract neurons, presumably by increased proliferation of myelin precursor cells, and thereby improves functional recovery. Most importantly, our study presents myelin loss as a novel therapeutic target following stroke.
    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
    半乳糖基-神经酰胺酶(GALC)是一种普遍存在的溶酶体酶,对于出生后早期发育期间哺乳动物神经系统的正确髓鞘形成至关重要。然而,成人大脑中GALC缺乏的生理后果仍然未知。在这项研究中,我们发现,在髓鞘化后少突胶质细胞中GALC活性有条件消除的小鼠在受到慢性实验性过敏性脑脊髓炎(EAE)攻击时被致命性致敏,与在无EAE攻击的GALC消融小鼠中观察到的非致死性髓鞘异常相反。机械上,我们发现,在TFEB依赖性溶酶体自噬体通量增加后,溶酶体和自噬体融合受损,髓鞘碎片积聚.这些结果表明,GALC缺乏的生理影响受细胞环境(少突胶质细胞vs全局表达)的高度影响,炎症的存在,以及发生时的发育时间(髓鞘形成前与髓鞘形成后)。我们得出的结论是,成人少突胶质细胞中的GALC表达对于维持成人中央髓鞘和减少对其他脱髓鞘损伤的脆弱性至关重要。
    Galactosyl-ceramidase (GALC) is a ubiquitous lysosomal enzyme crucial for the correct myelination of the mammalian nervous system during early postnatal development. However, the physiological consequence of GALC deficiency in the adult brain remains unknown. In this study, we found that mice with conditional ablation of GALC activity in post-myelinating oligodendrocytes were lethally sensitized when challenged with chronic experimental allergic encephalomyelitis (EAE), in contrast with the non-lethal dysmyelination observed in Galc-ablated mice without the EAE challenge. Mechanistically, we found strong inflammatory demyelination without remyelination and an impaired fusion of lysosomes and autophagosomes with accumulation of myelin debris after a transcription factor EB-dependent increase in the lysosomal autophagosome flux. These results indicate that the physiological impact of GALC deficiency is highly influenced by the cell context (oligodendroglial vs. global expression), the presence of inflammation, and the developmental time when it happens (pre-myelination vs. post-myelination). We conclude that Galc expression in adult oligodendrocytes is crucial for the maintenance of adult central myelin and to decrease vulnerability to additional demyelinating insults.
    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
    暂无摘要。
    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
    慢性脑低灌注可引起进行性脱髓鞘以及缺血性血管性痴呆,然而,没有有效的治疗方法。这里,基于脑白质损伤患者的磁共振成像研究,我们发现这种损伤与皮质结构紊乱有关.在老鼠模型中,在体感皮层中谷氨酸能神经元的光遗传学激活显着促进少突胶质祖细胞(OPC)的增殖,call体髓鞘再生,脑灌注不足后认知能力的恢复。这种刺激的治疗效果仅限于皮层的上层,但缺血后也跨越了很宽的时间窗。机械上,谷氨酸能神经元-OPC功能性突触连接的增强是实现激活皮质谷氨酸能神经元的保护作用所必需的。此外,皮肤抚摸,一种更容易转化为临床实践的方法,激活了体感皮层,从而促进OPC扩散,脑低灌注后的髓鞘再生和认知恢复。总之,我们证明,在慢性脑低灌注后,激活体感皮层中的谷氨酸能神经元促进OPCs的增殖和髓鞘再生以恢复认知功能。应该注意的是,这种激活可能通过精确调节谷氨酸能神经元-OPC电路为治疗缺血性血管性痴呆提供了新的方法。
    Chronic cerebral hypoperfusion can cause progressive demyelination as well as ischemic vascular dementia, however no effective treatments are available. Here, based on magnetic resonance imaging studies of patients with white matter damage, we found that this damage is associated with disorganized cortical structure. In a mouse model, optogenetic activation of glutamatergic neurons in the somatosensory cortex significantly promoted oligodendrocyte progenitor cell (OPC) proliferation, remyelination in the corpus callosum, and recovery of cognitive ability after cerebral hypoperfusion. The therapeutic effect of such stimulation was restricted to the upper layers of the cortex, but also spanned a wide time window after ischemia. Mechanistically, enhancement of glutamatergic neuron-OPC functional synaptic connections is required to achieve the protection effect of activating cortical glutamatergic neurons. Additionally, skin stroking, an easier method to translate into clinical practice, activated the somatosensory cortex, thereby promoting OPC proliferation, remyelination and cognitive recovery following cerebral hypoperfusion. In summary, we demonstrated that activating glutamatergic neurons in the somatosensory cortex promotes the proliferation of OPCs and remyelination to recover cognitive function after chronic cerebral hypoperfusion. It should be noted that this activation may provide new approaches for treating ischemic vascular dementia via the precise regulation of glutamatergic neuron-OPC circuits.
    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
    少突胶质细胞祖细胞(OPCs)是胶质细胞的一种亚型,产生少突胶质细胞,中枢神经系统(CNS)中的髓磷脂形成细胞。虽然OPC在发育过程中高度增殖,它们在成年期变得相对静止,当他们的命运受到细胞外环境的严格影响时。在外伤和慢性神经退行性疾病中,包括那些自身免疫起源的,少突胶质细胞经历凋亡,脱髓鞘开始了.成人OPCs立即激活;它们在病变部位迁移并增殖以补充受损区域,但是它们的效率受到神经胶质瘢痕的存在的阻碍,神经胶质瘢痕是主要由反应性星形胶质细胞形成的屏障,小胶质细胞和抑制性细胞外基质成分的沉积。如果,一方面,神经胶质疤痕限制了病变的扩散,它还会阻碍组织再生。已经提出了旨在减少星形胶质细胞或小胶质细胞激活并将其转变为神经保护表型的治疗策略。而OPC的作用在很大程度上被忽视了。在这次审查中,我们从OPCs的角度考虑了神经胶质疤痕,分析病变起源时的行为,并探索旨在维持OPCs以有效区分和促进髓鞘再生的潜在疗法。
    Oligodendrocyte progenitor cells (OPCs) represent a subtype of glia, giving rise to oligodendrocytes, the myelin-forming cells in the central nervous system (CNS). While OPCs are highly proliferative during development, they become relatively quiescent during adulthood, when their fate is strictly influenced by the extracellular context. In traumatic injuries and chronic neurodegenerative conditions, including those of autoimmune origin, oligodendrocytes undergo apoptosis, and demyelination starts. Adult OPCs become immediately activated; they migrate at the lesion site and proliferate to replenish the damaged area, but their efficiency is hampered by the presence of a glial scar-a barrier mainly formed by reactive astrocytes, microglia and the deposition of inhibitory extracellular matrix components. If, on the one hand, a glial scar limits the lesion spreading, it also blocks tissue regeneration. Therapeutic strategies aimed at reducing astrocyte or microglia activation and shifting them toward a neuroprotective phenotype have been proposed, whereas the role of OPCs has been largely overlooked. In this review, we have considered the glial scar from the perspective of OPCs, analysing their behaviour when lesions originate and exploring the potential therapies aimed at sustaining OPCs to efficiently differentiate and promote remyelination.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号