首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 137 毫秒
1.
It has recently been shown that bone marrow cells can differentiate into various lineage cells including neural cells in vitro and in vivo. We therefore examined whether bone marrow stem cells can differentiate into retinal neural cells in adult rats. PKH-67-labeled stem cell-enriched bone marrow cells (BMCs) were injected into the vitreous space of eyes in which the retinas had been mechanically injured using a hooked needle. Two weeks after the injection of these cells, immunohistochemical examinations were carried out. The stem cell-enriched BMCs had been incorporated and had differentiated into retinal neural cells in the injured retina. The stem cell-enriched BMCs had accumulated mainly in the outer nuclear layer around the injured sites. The incorporated cells expressed glial fibrillary acidic protein, calbindin, rhodopsin, and vimentin. These results raise the possibility that stem cell-enriched BMCs have the ability to differentiate into retinal neural cells, and that the injection of stem cell-enriched BMCs into the retina would help repair damaged retinal cells.  相似文献   

2.
Extensive research has focused on transplantation of pluripotent stem cells for the treatment of central nervous system disorders, the therapeutic potential of stem cell therapy for injured peripheral nerves is largely unknown. We used a rat sciatic nerve transection model to test the ability of implanted embryonic stem (ES) cell-derived neural progenitor cells (ES-NPCs) in promoting repair of a severely injured peripheral nerve. Mouse ES cells were neurally induced in vitro; enhanced expression and/or secretion of growth factors were detected in differentiating ES cells. One hour after removal of a 1-cm segment of the left sciatic nerve, ES-NPCs were implanted into the gap between the nerve stumps with the surrounding epineurium as a natural conduit. The transplantation resulted in substantial axonal regrowth and nerve repair, which were not seen in culture medium controls. One to 3 months after axotomy, co-immunostaining with the mouse neural cell membrane specific antibody M2/M6 and the Schwann cell marker S100 suggested that transplanted ES-NPCs had survived and differentiated into myelinating cells. Regenerated axons were myelinated and showed a uniform connection between proximal and distal stumps. Nerve stumps had near normal diameter with longitudinally oriented, densely packed Schwann cell-like phenotype. Fluoro-Gold retrogradely labeled neurons were found in the spinal cord (T12-13) and DRG (L4-L6), suggesting reconnection of axons across the transection. Electrophysiological recordings showed functional activity recovered across the injury gap. These data suggest that transplanted neurally induced ES cells differentiate into myelin-forming cells and provide a potential therapy for severely injured peripheral nerves.  相似文献   

3.
Mesenchymal stem cells (MSCs) were evaluated as an alternative source for tissue engineering of peripheral nerves. MSCs, transdifferentiated MSCs, or Schwann cells cultured from male rats were grafted into devitalized autologous muscle conduits bridging a 2-cm sciatic nerve gap in female rats. The differentiation potential of MSCs and transformed cultivated MSCs into Schwann cell-like cells was exploited using a cocktail of cytokines. Polymerase chain reaction of the SRY gene confirmed the presence of the implanted cells in the grafts. After 6 weeks, regeneration was monitored clinically, histologically, and morphometrically. Autologous nerves and cell-free muscle grafts were used as control. Revascularization studies suggested that transdifferentiated MSCs, in contrast to undifferentiated MSCs, facilitated neo-angiogenesis and did not influence macrophage recruitment. Autologous nerve grafts demonstrated the best results in all regenerative parameters. An appropriate regeneration was noted in the Schwann cell-groups and, albeit with restrictions, in the transdifferentiated MSC groups, whereas regeneration in the MSC group and in the cell-free group was impaired. The results indicate that transdifferentiated MSCs implanted into devitalized muscle grafts are able to support peripheral nerve regeneration to some extent, and offer a potential for new therapeutic strategies.  相似文献   

4.
In contrast to the peripheral nervous system (PNS), little structural and functional regeneration of the central nervous system (CNS) occurs spontaneously following injury in adult mammals. The inability of the CNS to regenerate is mainly attributed to its own inhibitorial environment such as glial scar formation and the myelin sheath of oligodendrocytes. Therefore, one of the strategies to promote axonal regeneration of the CNS is to experimentally modify the environment to be similar to that of the PNS. Schwann cells are the myelinating glial cells in the PNS, and are known to play a key role in Wallerian degeneration and subsequent regeneration. Central nervous system regeneration can be elicited by Schwann cell transplantation, which provides a suitable environment for regeneration. The underlying cellular mechanism of regeneration is based upon the cooperative interactions between axons and Schwann cells involving the production of neurotrophic factors and other related molecules. Furthermore, tight and gap junctional contact between the axon and Schwann cell also mediates the molecular interaction and linking. In this review, the role of the Schwann cell during the regeneration of the sciatic (representing the PNS) and optic (representing the CNS) nerves is explained. In addition, the possibility of optic nerve reconstruction by an artificial graft of Schwann cells is also described. Finally, the application of cells not of neuronal lineage, such as bone marrow stromal cells (MSCs), in nerve regeneration is proposed. Marrow stromal cells are known as multipotential stem cells that, under specific conditions, differentiate into several kinds of cells. The strategy to transdifferentiate MSCs into the cells with a Schwann cell phenotype and the induction of sciatic and optic nerve regeneration are described.  相似文献   

5.
Wang Y  Zhao Z  Ren Z  Zhao B  Zhang L  Chen J  Xu W  Lu S  Zhao Q  Peng J 《Neuroscience letters》2012,514(1):96-101
Chemical-extracted acellular nerve allografting, containing the natural nerve structure and elementary nerve extracellular matrix (ECM), has been used for peripheral nerve-defect treatment experimentally and clinically. However, functional outcome with acellular nerve allografting decreases with increased size of gap in nerve defects. Cell-based therapy is a good strategy for repairing long nerve defects. Bone-marrow-derived mesenchymal stem cells (BMSCs) and adipose-derived mesenchymal stem cells (ADSCs) can be induced to differentiate into cells with Schwann cell-like properties (BMSC-SCs or ADSC-SCs), which have myelin-forming ability in vitro and secrete trophic nerve growth factors. Here, we aimed to determine whether BMSC-SCs or ADSC-SCs are a promising cell type for enriching acellular grafts in nerve repair. We evaluated axonal regeneration distance by immunofluorescence staining after 2-week implantation. We used functional and histomorphometric analysis to evaluate 3-month regeneration of the novel cell-supplemented tissue-engineered nerve graft used to bridge a 15-mm-long sciatic nerve gap in rats. Introducing BMSC-SCs or ADSC-SCs to the acellular nerve graft promoted sciatic nerve regeneration and functional recovery. Nerve regeneration with BMSC-SCs or ADSC-SCs was comparable to that with autografting and Schwann cells alone and better than that with acellular nerve allografting alone. Differentiated bone-marrow-or adipose-derived MSCs may be a promising cell source for tissue-engineered nerve grafts and promote functional recovery after peripheral nerve injury.  相似文献   

6.
It has been reported that mesenchymal stem cells (MSCs) can transdifferentiate into Schwann cell-like cells by a series of treatments with a reducing agent, retinoic acid and a combination of trophic factors in vitro, and can transdifferentiate into myelin-forming cells to repair the demyelinated rat spinal cord in vivo. We now report that when co-cultured with dorsal root ganglion (DRG) neurons, MSCs were induced to transdifferentiate into Schwann cell-like cells that had ensheathed DRG axons. Following differentiation, MSCs underwent morphological changes similar to those of cultured Schwann cells and express GFAP and S100, the marker of Schwann cells. Moreover, 6 weeks later, MSCs wrapped their membrane around DRG axons. Further, initiation of myelination was observed in the co-cultured DRG neurons, which was determined by signals to MBP and this initiation of axon myelination by MSCs is similar to that of Schwann cells. However, electron micrographs show that no compact myelin was present in the MSCs co-cultures, whereas the Schwann cells co-cultures had formed a multilammelar myelin sheath around the axon. These indicate that the release of cytokine by DRG neurons may promote the transdifferentiation of MSCs, but is not sufficient to elicit compact myelination by transdifferentiated MSCs. These results improve our understanding in the mechanism of MSC transdifferentiation, and the mechanism underlying ensheathment and myelination by transdifferentiated MSCs.  相似文献   

7.
Peripheral nerves have the potential to regenerate axons and reinnervate end organs. Chronic denervation and disturbed nerve regeneration are thought to contribute to peripheral neuropathy, pain, and pruritus in the skin. The capacity of denervated distal nerves to support axonal regeneration requires proliferation by Schwann cells, which guide regenerating axons to their denervated targets. However, adult peripheral nerve Schwann cells do not retain a growth-permissive phenotype, as is required to produce new glia. Therefore, it is believed that following injury, mature Schwann cells dedifferentiate to a progenitor/stem cell phenotype to promote axonal regrowth. In this study, we show that skin-derived precursors (SKPs), a recently identified neural crest-related stem cell population in the dermis of skin, are an alternative source of progenitors for cutaneous nerve regeneration. Using in vivo and in vitro three-dimensional cutaneous nerve regeneration models, we show that the SKPs are neurotropic toward injured nerves and that they have a full capacity to differentiate into Schwann cells and promote axon regeneration. The identification of SKPs as a physiologic source of progenitors for cutaneous nerve regeneration in the skin, where SKPs physiologically reside, has important implications for understanding early cellular events in peripheral nerve regeneration. It also provides fertile ground for the elucidation of intrinsic and extrinsic factors within the nerve microenvironment that likely play essential roles in cutaneous nerve homeostasis. STEM Cells2012;30:2261-2270.  相似文献   

8.
背景:毛囊干细胞具有多分化潜能,可分化成神经细胞,极有希望成为治疗周围神经损伤的种子细胞。 目的:观察毛囊干细胞对坐骨神经损伤修复的影响。 方法:体外分离培养SD大鼠乳鼠胡须处的毛囊干细胞,经鉴定备用。36只SD大鼠随机分为实验组和对照组,建立坐骨神经损伤模型后,实验组于坐骨神经损伤处的上方注入浓度约106 L-1的毛囊干细胞50 μL,对照组注射等量的磷酸盐缓冲液。 结果与结论:各组坐骨神经功能指数均随观察时间进行性增加,其中实验组大鼠神经功能恢复早于对照组;免疫组织化学检测移植后的毛囊干细胞大量存活并分化成神经细胞。结果提示毛囊干细胞能够有效的促进损伤的坐骨神经修复。  相似文献   

9.
Adult peripheral nerves in vertebrates can regrow their axons and re-establish function after crush lesion. However, when there is extensive loss of a nerve segment, due to an accident or compressive damage caused by tumors, regeneration is strongly impaired. In order to overcome this problem, bioengineering strategies have been employed, using biomaterials formed by key cell types combined with biodegradable polymers. Many of these strategies are successful, and regenerated nerve tissue can be observed 12 weeks after the implantation. Mesenchymal stem cells (MSCs) are one of the key cell types and the main stem-cell population experimentally employed for cell therapy and tissue engineering of peripheral nerves. The ability of these cells to release a range of different small molecules, such as neurotrophins, growth factors and interleukins, has been widely described and is a feasible explanation for the improvement of nerve regeneration. Moreover, the multipotent capacity of MSCs has been very often challenged with demonstrations of pluripotency, which includes differentiation into any neural cell type. In this study, we generated a biomaterial formed by EGFP-MSCs, constitutively covering microstructured filaments made of poly-ε-caprolactone. This biomaterial was implanted in the sciatic nerve of adult rats, replacing a 12-mm segment, inside a silicon tube. Our results showed that six weeks after implantation, the MSCs had differentiated into connective-tissue cells, but not into neural crest-derived cells such as Schwann cells. Together, present findings demonstrated that MSCs can contribute to nerve-tissue regeneration, producing trophic factors and differentiating into fibroblasts, endothelial and smooth-muscle cells, which compose the connective tissue.  相似文献   

10.
Ao Q  Fung CK  Tsui AY  Cai S  Zuo HC  Chan YS  Shum DK 《Biomaterials》2011,32(3):787-796
Autologous nerve grafts have been the 'gold standard' for treatment of peripheral nerve defects that exceed the critical gap length. To address issues of limited availability of donor nerves and donor site morbidity, we have fabricated chitosan conduits and seeded them with bone marrow stromal cell (BMSC)-derived Schwann cells as an alternative. The derived Schwann cells used were checked for fate commitment. The conduits were tested for efficacy in bridging the critical gap length of 12 mm in sciatic nerves of adult rats. By three months post-operation, mid-shank circumference, nerve conduction velocity, average regenerated myelin area, and myelinated axon count, in nerves bridged with BMSC-derived Schwann cells were similar to those treated with sciatic nerve-derived Schwann cells (p > 0.05) but significantly higher than those bridged with PBS-filled conduits (p < 0.05). Evidence is thus provided in support of the use of chitosan conduits seeded with BMSC-derived Schwann cells to treat critical defects in peripheral nerves. This provides the basis to pursue BMSC as an autologous source of Schwann cells for transplantation therapy in larger animal species.  相似文献   

11.
Mesenchymal stem cells (MSCs) represent a promising therapeutic approach in nerve tissue engineering. To date, the local implantation of MSC in injured nerves has been the only route of administration used. In case of multiple sites of injury, the systemic administration of cells capable of reaching damaged nerves would be advisable. In this regard, we found that an intravenous administration of adipose-derived MSC (ASC) 1 week after sciatic nerve crush injury, a murine model of acute axonal damage, significantly accelerated the functional recovery. Sciatic nerves from ASC-treated mice showed the presence of a restricted number of undifferentiated ASC together with a significant improvement in fiber sprouting and the reduction of inflammatory infiltrates for up to 3 weeks. Besides the immune modulatory effect, our results show that ASC may contribute to peripheral nerve regeneration because of their ability to produce in culture neuroprotective factors such as insulin-like growth factor I, brain-derived neurotrophic factor, or basic fibroblast growth factor. In addition to this production in vitro, we interestingly found that the concentration of glial-derived neurotrophic factor (GDNF) was significantly increased in the sciatic nerves in mice treated with ASC. Since no detectable levels of GDNF were observed in ASC cultures, we hypothesize that ASC induced the local production of GDNF by Schwann cells. In conclusion, we show that systemically injected ASC have a clear therapeutic potential in an acute model of axonal damage. Among the possible mechanisms promoting nerve regeneration, our results rule out a process of trans-differentiation and rather suggest the relevance of a bystander effect, including the production of in situ molecules, which, directly or indirectly through a cross-talk with local glial cells, may modulate the local environment with the down-regulation of inflammation and the promotion of axonal regeneration.  相似文献   

12.
Hou SY  Zhang HY  Quan DP  Liu XL  Zhu JK 《Neuroscience》2006,140(1):101-110
Bone marrow stromal cells are multipotential stem cells that contribute to the differentiation of tissues such as bone, cartilage, fat and muscle. In the experiment, we found that bone marrow stromal cells can be induced to differentiate into cells expressing characteristic markers of Schwann cells, such as S-100 and glial fibrillary acidic protein, promoting peripheral nerve regeneration. Tissue-engineered bioartificial nerve grafting of rats by differentiated bone marrow stromal cells was applied for bridging a 10 mm-long sciatic nerve defect. Twenty-eight inbred strains of female F344 rats weighing 160 approximately 200 g were randomly divided into four nerve grafting groups, with seven rats in each group. Differentiated bone marrow stromal cell-laden group: poly(lactic-co-glycolic) acid tubes with an intrinsic framework were seeded with syngeneic bone marrow stromal cells which were induced for 5 days; Schwann cell-laden group: poly(lactic-co-glycolic) acid tubes with an intrinsic framework were seeded with syngeneic Schwann cells; acellular group: poly(lactic-co-glycolic) acid tubes were only filled with an intrinsic framework; autografts group. Three months later, a series of examinations was performed, including electrophysiological methods, walking track analysis, immunohistological staining of nerves, immunostaining of S-100 and neurofilament, and axon counts. The outcome indicated that bone marrow stromal cells are able to differentiate into Schwann-like cells and Schwann-like cells could promote nerve regeneration. Bone marrow stromal cells may be potentially optional seed cells for peripheral nerve tissue engineering because of abilities of promoting axonal regeneration.  相似文献   

13.
Despite the fact that the peripheral nervous system is able to regenerate after traumatic injury, the functional outcomes following damage are limited and poor. Bone marrow mesenchymal stem cells (MSCs) are multipotent cells that have been used in studies of peripheral nerve regeneration and have yielded promising results. The aim of this study was to evaluate sciatic nerve regeneration and neuronal survival in mice after nerve transection followed by MSC treatment into a polycaprolactone (PCL) nerve guide. The left sciatic nerve of C57BL/6 mice was transected and the nerve stumps were placed into a biodegradable PCL tube leaving a 3-mm gap between them; the tube was filled with MSCs obtained from GFP+ animals (MSC-treated group) or with a culture medium (Dulbecco's modified Eagle's medium group). Motor function was analyzed according to the sciatic functional index (SFI). After 6 weeks, animals were euthanized, and the regenerated sciatic nerve, the dorsal root ganglion (DRG), the spinal cord, and the gastrocnemius muscle were collected and processed for light and electron microscopy. A quantitative analysis of regenerated nerves showed a significant increase in the number of myelinated fibers in the group that received, within the nerve guide, stem cells. The number of neurons in the DRG was significantly higher in the MSC-treated group, while there was no difference in the number of motor neurons in the spinal cord. We also found higher values of trophic factors expression in MSC-treated groups, especially a nerve growth factor. The SFI revealed a significant improvement in the MSC-treated group. The gastrocnemius muscle showed an increase in weight and in the levels of creatine phosphokinase enzyme, suggesting an improvement of reinnervation and activity in animals that received MSCs. Immunohistochemistry documented that some GFP+ -transplanted cells assumed a Schwann-cell-like phenotype, as evidenced by their expression of the S-100 protein, a Schwann cell marker. Our findings suggest that using a PCL tube filled with MSCs is a good strategy to improve nerve regeneration after a nerve transection in mice.  相似文献   

14.
大鼠骨髓间质细胞向Schwann-like细胞的诱导分化   总被引:1,自引:0,他引:1  
目的探索成年大鼠骨髓间质细胞(MSCs)向Schwann—like细胞转化的方法和机理。方法分别用变性神经提取液和含有大鼠白血病抑制因子(rLIF)、上皮生长因子(EGF)、碱性成纤维细胞生长因子(bFGF)等的诱导液诱导成年大鼠骨髓间质细胞,观察诱导过程中细胞形态学变化,利用免疫细胞化学染色鉴定诱导细胞性质;同时培养大鼠Schwann细胞作为对照。结果经变性神经提取液诱导后细胞转化为梭形细胞,并排列成网状;经细胞因子诱导后细胞呈梭形排列,S-100,GFAP染色呈阳性反应。结论变性神经提取液中含有促使骨髓间质细胞向Schwann-like细胞分化的必要成份;LIF可能是这些必要成份的重要组成之一。  相似文献   

15.
目的研究重组睫状神经营养因子(CNTF)对受损周围神经施万细胞基因表达的作用。方法用硅管套接切断的大鼠坐骨神经,在受损神经局部给予重组CNTF,术后用免疫组织化学ABC法结合计算机图像分析观测S100蛋白(S100)、生长相关蛋白-43(GAP-43)、磷酸化酪氨酸(PTyr)、信号转导子和转录激活子(STAT)3的免疫反应阳性物质在修复侧远段神经的分布和相对含量。结果CNTF组修复侧远段神经相应区域S100、GAP-43、PTyr、STAT3阳性物质的含量显著或非常显著高于生理盐水组。结论重组CNTF能上调受损神经施万细胞S100、GAP-43、PTyr和STAT3的表达,提示重组CNTF通过强化受损神经施万细胞JAK-STAT途径,E调其S100和GAP-43的基因表达。  相似文献   

16.
Immortalized rat Schwann cells produce tumoursin vivo   总被引:3,自引:0,他引:3  
Summary We have recently reported the immortalization of primary Schwann cells isolated from sciatic nerves of normal neonatal rats. The cells were maintained under continuous mitogenic stimulation with glial growth factor and forskolin, achieving immortalization after 12 to 15 weeks without the use of viral infection, oncogene transformation or chemical carcinogens. The immortalized cells (1.17 cells) initially retain the capability to recognize and attach to peripheral neurons in culture as well as the ability to myelinate those neurons. The functional capacity of the cells gradually diminishes in culture, such that late passage cells can ensheath neurons but cannot form a myelin sheath. Both normal and immortalized cells secrete comparable amounts of autocrine growth factor activity in culture that can be regulated by extracellular matrix proteins. The difference between quiescent and immortalized Schwann cells seems to lie not in the production of growth factor but rather in the relative ability to respond to the factor(s). To test the potential of the immortalized Schwann cells for the ability to form tumoursin vivo, we injected equal numbers of primary or immortalized Schwann cells into the sciatic nerve of adult syngenic rats and allowed them to incubate there for 6 to 13 weeks, whereupon the injected nerves were inspected for tumour formation. In every case (N=3) the primary cells had no effect whereas every injection of immortalized cells (N=5) resulted in a solid cellular mass surrounding the injected nerve. The tumours were encapsulated masses of actively dividing Schwann-like cells that surrounded but did not invade the nerve fascicle. The cells in the tumour expressed the Schwann cell surface antigens laminin, 217C (Ran 1) and S-100 like the immortalized cells that had been injected. Within the tumour the cells were embedded in a collagenous matrix, were surrounded by basal lamina and occasionally attained an orientation comparable to the Antoni A or Antoni B patterns typical of human schwannomas. These data suggest that rat Schwann cells immortalizedin vitro by chronic mitogenic stimulation can provide an experimental animal model for human schwannomas and neurofibromas.  相似文献   

17.
脱细胞同种异体神经移植物的制备及成分分析   总被引:7,自引:2,他引:7  
目的 探讨脱细胞同种异体神经移植的制备方法及其含有的成分。方法 采用组织工程化低渗脱细胞方法制备神经异体移植物 ,光电镜观察其结构特征 ;用免疫组织化学法和聚丙烯酰胺凝胶电泳法分析神经异体移植物的成分。结果 正常的 (天然的 )周围神经经脱细胞处理后 ,清除了雪旺细胞、神经外膜和束膜的细胞 ,以及神经纤维的髓鞘和轴突 ,保存了由雪旺细胞基底膜管以及神经外膜和束膜的细胞外基质构成的三维支架结构。成分分析结果显示 ,含有LN、FN以及生长相关蛋白GAP 4 3和 6 5kDa蛋白等促进和诱导受体损伤神经再生的重要成分。结论 脱细胞异体神经移植物含有诱导和促进神经再生的相关蛋白 ,有利于受损神经的再生。  相似文献   

18.
The effects of addition of Schwann cells on peripheral nerve regeneration through a novel graft material-the tendon autograft-and a conventional freeze-thawed muscle graft, were studied in the rat sciatic nerve. Adult Schwann cell cultures were established from predegenerated nerves. The Schwann cells were added to the autologous grafts by coculture (tendon autograft) or injection (freeze-thawed muscle graft). Both graft types supported adherence of the added Schwann cells. Addition of cultured Schwann cells to the two different graft models improved regeneration by increasing the rate of axonal outgrowth as compared with similar grafts without added cells.  相似文献   

19.
Summary We have conducted experiments in the adult rat visual system to assess the relative importance of an absence of trophic factors versus the presence of putative growth inhibitory molecules for the failure of regeneration of CNS axons after injury. The experiments comprised three groups of animals in which all optic nerves were crushed intra-orbitally: an optic nerve crush group had a sham implant-operation on the eye; the other two groups had peripheral nerve tissue introduced into the vitreous body; in an acellular peripheral nerve group, a frozen/thawed teased sciatic nerve segment was grafted, and in a cellular peripheral nerve group, a predegenerate teased segment of sciatic nerve was implanted. The rats were left for 20 days and their optic nerves and retinae prepared for immunohistochemical examination of both the reaction to injury of axons and glia in the nerve and also the viability of Schwann cells in the grafts. Anterograde axon tracing with rhodamine-B provided unequivocal qualitative evidence of regeneration in each group, and retrograde HRP tracing gave a measure of the numbers of axons growing across the lesion by counting HRP filled retinal ganglion cells in retinal whole mounts after HRP injection into the optic nerve distal to the lesion. No fibres crossed the lesion in the optic nerve crush group and dense scar tissue was formed in the wound site. GAP-43-positive and rhodamine-B filled axons in the acellular peripheral nerve and cellular peripheral nerve groups traversed the lesion and grew distally. There were greater numbers of regenerating fibres in the cellular peripheral nerve compared to the acellular peripheral nerve group. In the former, 0.6–10% of the retinal ganglion cell population regenerated axons at least 3–4 mm into the distal segment. In both the acellular peripheral nerve and cellular peripheral nerve groups, no basal lamina was deposited in the wound. Thus, although astrocyte processes were stacked around the lesion edge, a glia limitans was not formed. These observations suggest that regenerating fibres may interfere with scarring. Viable Schwann cells were found in the vitreal grafts in the cellular peripheral nerve group only, supporting the proposition that Schwann cell derived trophic molecules secreted into the vitreous stimulated retinal ganglion cell axon growth in the severed optic nerve. The regenerative response of acellular peripheral nerve-transplanted animals was probably promoted by residual amounts of these molecules present in the transplants after freezing and thawing. In the optic nerves of all groups the astrocyte, microglia and macrophage reactions were similar. Moreover, oligodendrocytes and myelin debris were also uniformly distributed throughout all nerves. Our results suggest either that none of the above elements inhibit CNS regeneration after perineuronal neurotrophin delivery, or that the latter, in addition to mobilising and maintaining regeneration, also down regulates the expression of axonal growth cone-located receptors, which normally mediate growth arrest by engaging putative growth inhibitory molecules of the CNS neuropil.  相似文献   

20.
Summary Injury of the peripheral axons of primary sensory neurons has been previously shown to increase the probability that the corresponding central axons would grow from the injured spinal cord into a peripheral nerve graft. This phenomenon has been used to investigate the nature of extrinsic cues from injured nerves that trigger an enhanced regenerative propensity within sensory neurons. In 13 groups of rats, a segment of the right sciatic nerve was grafted to the dorsal columns of the spinal cord and the left sciatic nerve was subjected to mechanical injury, injection of colchicine or infusion of nerve growth factor. Subsequently, neurons in lumbar dorsal root ganglia with axons growing from the spinal cord into a graft were identified by retrograde perikaryal labelling and compared for the two sides. The aim was to mimic or modify the inductive effect of nerve transaction by alternative or additional manipulation of the nerve. Growth of central axons was less enhanced by peripheral axonal interruption if the length of the proximal stump was increased or if a distal stump was present to permit rapid regeneration. However, the regenerative response following nerve transection was altered little by crushing the proximal stump or injecting it with colchicine or nerve growth factor. It is suggested that sensory neurons are stimulated to regenerate by peripheral axonal injuries that reduce some normal retrograde regulatory influence of Schwann cells.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号