remyelination

髓鞘再生
  • 文章类型: Journal Article
    神经可塑性作为克服由不同神经系统疾病引起的中枢神经系统损伤的机制近年来受到越来越多的关注。然而,这些修复机制的缺乏导致神经元损伤的积累,因此长期残疾。迄今为止,髓鞘再生发生的机制以及为何髓鞘再生的程度在多发性硬化患者之间不同而与病程无关,目前尚不清楚.神经营养因子家族的一员,脑源性神经营养因子(BDNF)在这种情况下受到特别关注,因为它被认为在髓鞘再生和神经可塑性中起着核心作用,神经保护,和记忆。
    分析当前有关多发性硬化症不同领域BDNF的文献,并概述该领域的知识现状。
    到目前为止,评估BDNF在多发性硬化症患者中的作用的研究仍然没有定论.然而,有新的证据表明BDNF对多发性硬化症有有益的作用,因为报告对临床和MRI特征有积极影响的研究超过了假设BDNF有害影响的研究。此外,关于Val66Met多态性的研究尚未最终确定这是多发性硬化症的保护性因素还是有害因素,但大多数研究再次假设通过调节BDNF分泌和抗炎作用的保护作用,在健康对照和多发性硬化症患者中具有不同的作用,可能是由于多发性硬化症患者的促炎环境。需要进行更大的队列和纵向随访的进一步研究,以提高我们对BDNF在中枢神经系统中的作用的理解,尤其是在多发性硬化症的背景下。
    UNASSIGNED: Neuroplasticity as a mechanism to overcome central nervous system injury resulting from different neurological diseases has gained increasing attention in recent years. However, deficiency of these repair mechanisms leads to the accumulation of neuronal damage and therefore long-term disability. To date, the mechanisms by which remyelination occurs and why the extent of remyelination differs interindividually between multiple sclerosis patients regardless of the disease course are unclear. A member of the neurotrophins family, the brain-derived neurotrophic factor (BDNF) has received particular attention in this context as it is thought to play a central role in remyelination and thus neuroplasticity, neuroprotection, and memory.
    UNASSIGNED: To analyse the current literature regarding BDNF in different areas of multiple sclerosis and to provide an overview of the current state of knowledge in this field.
    UNASSIGNED: To date, studies assessing the role of BDNF in patients with multiple sclerosis remain inconclusive. However, there is emerging evidence for a beneficial effect of BDNF in multiple sclerosis, as studies reporting positive effects on clinical as well as MRI characteristics outweighed studies assuming detrimental effects of BDNF. Furthermore, studies regarding the Val66Met polymorphism have not conclusively determined whether this is a protective or harmful factor in multiple sclerosis, but again most studies hypothesized a protective effect through modulation of BDNF secretion and anti-inflammatory effects with different effects in healthy controls and patients with multiple sclerosis, possibly due to the pro-inflammatory milieu in patients with multiple sclerosis. Further studies with larger cohorts and longitudinal follow-ups are needed to improve our understanding of the effects of BDNF in the central nervous system, especially in the context of multiple sclerosis.
    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
    背景:过度的神经炎症,凋亡,胶质疤痕,脊髓损伤(SCI)引发的脱髓鞘是SCI修复的主要障碍。褐藻多糖,天然海洋植物提取物,具有广谱抗炎和免疫调节作用,被认为是治疗各种疾病的潜在药物,包括神经系统疾病.然而,其在SCI中的作用尚未得到调查。
    方法:在本研究中,我们在小鼠中建立了SCI模型,并通过每天腹膜内注射不同剂量的岩藻依聚糖(10和20mg/kg)来干预损伤修复。同时,体外处理原代少突胶质前体细胞(OPCs)以验证岩藻依聚糖对OPCs的分化促进作用。Basso鼠标秤(BMS),路易斯维尔游泳量表(LSS),进行了旋转试验以测量功能恢复。免疫荧光染色,和透射电子显微镜(TEM)进行评估神经炎症,凋亡,胶质疤痕,和髓鞘再生。进行Western印迹分析以阐明髓鞘再生的潜在机制。
    结果:我们的结果表明,在SCI模型中,岩藻依聚糖表现出显著的抗炎作用,并促进促炎M1型小胶质细胞/巨噬细胞向抗炎M2型的转化。岩藻依聚糖增强损伤区域的神经元和轴突的存活并改善髓鞘再生。此外,岩藻依聚糖通过激活PI3K/AKT/mTOR通路促进OPCs分化为成熟少突胶质细胞。
    结论:岩藻依聚糖通过调节微环境和促进髓鞘再生来改善SCI修复。
    BACKGROUND: 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.
    CONCLUSIONS: Fucoidan improves SCI repair by modulating the microenvironment and promoting remyelination.
    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
    多发性硬化(MS)是一种导致炎性脱髓鞘的慢性和使人衰弱的神经系统疾病。虽然内源性髓鞘再生有助于恢复功能,随着时间的推移,这种恢复过程的效率往往会降低。目前,针对促进髓鞘再生的机制的努力被认为是有希望的治疗方法。M1毒蕈碱乙酰胆碱受体(M1R)先前被鉴定为少突胶质细胞分化和髓鞘形成的负调节剂。这里,我们通过使用高选择性M1R探针表征人和啮齿动物少突胶质细胞(包括人MS组织中的细胞)中的表达,验证M1R是髓鞘再生的靶标.作为传统方法论的突破,我们将荧光团与高度M1R选择性肽(MT7)缀合,该肽靶向亚纳摩尔范围的M1R。这允许异常检测人CNS中的M1R蛋白表达。更重要的是,我们引入PIPE-307,一种脑渗透剂,具有良好的药物样特性的小分子拮抗剂,选择性靶向M1R。我们在一系列体外和体内研究中评估了PIPE-307,以表征M1R相对于M2-5R的效力和选择性,并确认了阻断该受体以促进分化和髓鞘再生的充分性。Further,PIPE-307在MS的小鼠实验性自身免疫性脑脊髓炎模型中显示出显着疗效,组织学,电子显微镜,和视觉诱发电位。一起,这些发现支持靶向M1R用于髓鞘再生,并支持PIPE-307的进一步开发用于临床研究.
    Multiple sclerosis (MS) is a chronic and debilitating neurological disease that results in inflammatory demyelination. While endogenous remyelination helps to recover function, this restorative process tends to become less efficient over time. Currently, intense efforts aimed at the mechanisms that promote remyelination are being considered promising therapeutic approaches. The M1 muscarinic acetylcholine receptor (M1R) was previously identified as a negative regulator of oligodendrocyte differentiation and myelination. Here, we validate M1R as a target for remyelination by characterizing expression in human and rodent oligodendroglial cells (including those in human MS tissue) using a highly selective M1R probe. As a breakthrough to conventional methodology, we conjugated a fluorophore to a highly M1R selective peptide (MT7) which targets the M1R in the subnanomolar range. This allows for exceptional detection of M1R protein expression in the human CNS. More importantly, we introduce PIPE-307, a brain-penetrant, small-molecule antagonist with favorable drug-like properties that selectively targets M1R. We evaluate PIPE-307 in a series of in vitro and in vivo studies to characterize potency and selectivity for M1R over M2-5R and confirm the sufficiency of blocking this receptor to promote differentiation and remyelination. Further, PIPE-307 displays significant efficacy in the mouse experimental autoimmune encephalomyelitis model of MS as evaluated by quantifying disability, histology, electron microscopy, and visual evoked potentials. Together, these findings support targeting M1R for remyelination and support further development of PIPE-307 for clinical studies.
    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
    白质损伤(WMI)被认为是创伤性脑损伤(TBI)后长期认知功能障碍的主要原因。这种损伤部分是由于损伤后少突胶质细胞谱系细胞(OLC)的凋亡死亡。由创伤直接触发或响应退化的轴突。最近的研究表明,肠道菌群通过调节TBI后外周免疫细胞浸润来调节炎症反应。此外,T细胞直接影响OLC分化和增殖。因此,我们假设肠道菌群在调节OLC对影响T细胞分化和活化的WMI反应中起关键作用.TBI后早期肠道微生物消耗慢性减少髓鞘再生,OLC增殖急剧减少,并与髓鞘碎片积累增加有关。令人惊讶的是,TBI后,肠道微生物群耗尽小鼠中T细胞缺失可恢复OLC增殖和髓鞘再生.与来自对照损伤小鼠的T细胞相比,与来自肠道微生物群耗尽小鼠的T细胞共培养的OLC导致增殖受损和MHC-II表达增加。此外,在肠道微生物群耗竭和TBI条件下,OLC中的MHC-II表达似乎与受损的增殖有关。总的来说,我们的数据表明,TBI后肠道微生物群的消耗受损,OLC增殖减少,同时OLCMHCII表达增加,并且需要T细胞的存在。该数据表明,T细胞是肠道微生物群调节TBI后少突胶质细胞反应和白质恢复的重要机制联系。
    White matter injury (WMI) is thought to be a major contributor to long-term cognitive dysfunctions after traumatic brain injury (TBI). This damage occurs partly due to apoptotic death of oligodendrocyte lineage cells (OLCs) after the injury, triggered directly by the trauma or in response to degenerating axons. Recent research suggests that the gut microbiota modulates the inflammatory response through the regulation of peripheral immune cell infiltration after TBI. Additionally, T-cells directly impact OLCs differentiation and proliferation. Therefore, we hypothesized that the gut microbiota plays a critical role in regulating the OLC response to WMI influencing T-cells differentiation and activation. Gut microbial depletion early after TBI chronically reduced re-myelination, acutely decreased OLCs proliferation, and was associated with increased myelin debris accumulation. Surprisingly, the absence of T-cells in gut microbiota depleted mice restored OLC proliferation and remyelination after TBI. OLCs co-cultured with T-cells derived from gut microbiota depleted mice resulted in impaired proliferation and increased expression of MHC-II compared with T cells from control-injured mice. Furthermore, MHC-II expression in OLCs appears to be linked to impaired proliferation under gut microbiota depletion and TBI conditions. Collectively our data indicates that depletion of the gut microbiota after TBI impaired remyelination, reduced OLCs proliferation with concomitantly increased OLC MHCII expression, and required the presence of T cells. This data suggests that T cells are an important mechanistic link by which the gut microbiota modulate the oligodendrocyte response and white matter recovery after TBI.
    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
    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

       PDF(Pubmed)

  • 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

       PDF(Pubmed)

  • 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
    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: Journal Article
    暂无摘要。
    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
    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)

公众号