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 15 毫秒
1.
Goldfish retinal ganglion cells (RGCs) can regrow their axons after optic nerve injury. However, the reason why goldfish RGCs can regenerate after nerve injury is largely unknown at the molecular level. To investigate regenerative properties of goldfish RGCs, we divided the RGC regeneration process into two components: (1) RGC survival, and (2) axonal elongation processes. To characterize the RGC survival signaling pathway after optic nerve injury, we investigated cell survival/death signals such as Bcl-2 family members in the goldfish retina. Amounts of phospho-Akt (p-Akt) and phospho-Bad (p-Bad) in the goldfish retina rapidly increased four- to five-fold at the protein level by 3-5 days after nerve injury. Subsequently, Bcl-2 levels increased 1.7-fold, accompanied by a slight reduction in caspase-3 activity 10-20 days after injury. Furthermore, level of insulin-like growth factor-I (IGF-I), which activates the phosphatidyl inositol-3-kinase (PI3K)/Akt system, increased 2-3 days earlier than that of p-Akt in the goldfish retina. The cellular localization of these molecular changes was limited to RGCs. IGF-I treatment significantly induced phosphorylation of Akt, and strikingly induced neurite outgrowth in the goldfish retina in vitro. On the contrary, addition of the PI3K inhibitor wortmannin, and IGF-I antibody inhibited Akt phosphorylation and neurite outgrowth in an explant culture. Thus, we demonstrated, for the first time, the signal cascade for early upregulation of IGF-I, leading to RGC survival and axonal regeneration in adult goldfish retinas through PI3K/Akt system after optic nerve injury. The present data strongly indicate that IGF-I is one of the most important molecules for controlling regeneration of RGCs after optic nerve injury.  相似文献   

2.
3.
4.
Autophagy is an essential recycling pathway implicated in neurodegeneration either as a pro-survival or a pro-death mechanism. Its role after axonal injury is still uncertain. Axotomy of the optic nerve is a classical model of neurodegeneration. It induces retinal ganglion cell death, a process also occurring in glaucoma and other optic neuropathies. We analyzed autophagy induction and cell survival following optic nerve transection (ONT) in mice. Our results demonstrate activation of autophagy shortly after axotomy with autophagosome formation, upregulation of the autophagy regulator Atg5 and apoptotic death of 50% of the retinal ganglion cells (RGCs) after 5 days. Genetic downregulation of autophagy using knockout mice for Atg4B (another regulator of autophagy) or with specific deletion of Atg5 in retinal ganglion cells, using the Atg5(flox/flox) mice reduces cell survival after ONT, whereas pharmacological induction of autophagy in vivo increases the number of surviving cells. In conclusion, our data support that autophagy has a cytoprotective role in RGCs after traumatic injury and may provide a new therapeutic strategy to ameliorate retinal diseases.  相似文献   

5.
Changes in retinal neuronal populations in the DBA/2J mouse   总被引:1,自引:0,他引:1  
DBA/2J (D2) mice develop a form of progressive pigmentary glaucoma with increasing age. We have compared retinal cell populations of D2 mice with those in control C57BL/6J mice to provide information on retinal histopathology in the D2 mouse. The D2 mouse retina is characterized by a reduction in retinal thickness caused mainly by a thinning of the inner retinal layers. Immunocytochemical staining for specific inner retinal neuronal markers, viz., calbindin for horizontal cells; protein kinase C (PKC) and recoverin for bipolar cells, glycine, -aminobutyric acid (GABA), choline acetyltransferase (ChAT), and nitric oxide synthase (NOS) for amacrine cells, and osteopontin (OPN) for ganglion cells, was performed to detect preferentially affected neurons in the D2 mouse retina. Calbindin, PKC, and recoverin immunoreactivities were not significantly altered. Amacrine cells immunoreactive for GABA, ChAT, and OPN were markedly decreased in number, whereas NOS-immunoreactive amacrine cells increased in number. However, no changes were observed in the population of glycine-immunoreactive amacrine cells. These findings indicate a significant loss of retinal ganglion and some amacrine cells, whereas glycinergic amacrine cells, horizontal, and bipolar cells are almost unaffected in the D2 mouse. The reduction in amacrine cells appears to be attributable to a loss of GABAergic and particularly cholinergic amacrine cells. The increase in nitrergic neurons with the consequent increase in NOS and NO may be important in the changes in the retinal organization that lead to glaucomain D2 mice. Thus, the D2 mouse retina represents a useful model for studying the pathogenesis of glaucoma and mechanisms of retinal neuronal death and for evaluating neuroprotection strategies.Jung-Il Moon and In-Beom Kim contributed equally to this work.This work was supported by a Korea Research Foundation Grant (FP 0005) and by BK 21 in Korea.  相似文献   

6.
7.
Cell cycle re-entry is one of the key processes in neuronal apoptosis. Previous studies have shown that Ski-interacting protein (SKIP) played an important role in cell cycle re-entry. However, its expression and function in optic nerve injury are still with limited acquaintance. To investigate whether SKIP is involved in retinal ganglion cells (RGCs) death, we performed an optic nerve crush (ONC) model in adult rats. Western blot analysis revealed that up-regulation of SKIP was present in retina at 5 days after ONC. Immunofluorescent labeling indicated that up-regulated SKIP was found mainly in RGCs. We also investigated co-localization of SKIP with active-caspase-3 and TUNEL (apoptotic markers) -positive cells in the retina after ONC. In addition, the expression of SKIP was increased in parallel with P53 and P21 in retina after ONC. All these results suggested that up-regulation of SKIP in the retina was associated with RGCs death after ONC.  相似文献   

8.
Retinal ganglion cells (RGCs) die by apoptosis after optic nerve injury. A number of reports have separately shown changes in pro-apoptotic proteins such as the Bcl-2 family members following optic nerve injury. However, induction time of these apoptotic signals has not been identified due to different treatments of the optic nerve, and insufficient time intervals for measurements. Therefore, the stream of cell death signals is not well understood. In the present study, we systematically reinvestigated a detailed time course of these cell death/survival signals in the rat retina after optic nerve crush, to determine the signal cascade leading to RGC apoptosis. The most conspicuous changes detected in the retina were the rapid inactivation of phospho-Akt and phospho-Bad proteins 2-3 days after optic nerve damage, and the subsequent gradual activation of Bax protein and caspase-3 activity accompanied by cell loss of RGCs 6 days after nerve injury. Cellular localization of these molecular changes was limited to RGCs. Furthermore, amount of insulin-like growth factor-I (IGF-I), an activator of the phosphatidyl inositol-3-kinase (PI3K)/Akt system, was initially decreased from RGCs 1-2 days just prior to the inactivation of phospho-Akt by optic nerve crush. Conversely, supplementation with IGF-I into the rat retina induced upregulation of phospho-Akt expression and cell survival of RGCs both in vitro and in vivo. Thus, injury to the optic nerve might induce early changes in cellular homeostasis with a plausible loss of trophic support for injured RGCs. Actually, IGF-I drastically enhanced neurite outgrowth from adult rat RGCs via a wortmannin-dependent mechanism in a retinal explant culture. Our data strongly indicate that IGF-I is a key molecule that induces RGC apoptosis or RGC survival and regeneration in the retina during the early stage of optic nerve injury.  相似文献   

9.
After sectioning of the goldfish optic nerve a number of enzyme histochemical changes are observed in the hypertrophied retinal ganglion cells and in the optic nerve. Between one and eighteen days postoperatively an increase in the amount of acid phosphatase reaction product is noted. The enhanced activity decreased to normal first in the optic nerve, followed by the optic tract and tectum. Four days postoperatively higher levels of activity were noted in the hypertrophic retinal ganglion cells for the enzymes NADH tetrazolium reductase, cytochrome oxidase, glutamate dehydrogenase and lactate dehydrogenase. The same enzymes also showed an activity increase in the lesioned optic nerve after four to ten days postoperatively, beginning at the cut and gradually spreading towards the optic tectum. Between fifteen and eighteen days the activity dropped to normal in the hypertrophic retinal ganglion cells, while in the lesioned nerve raised levels of reaction products could be seen till days thirty-five and/or forty-five. It was concluded that the degeneration of the optic pathway is marked by the increase of acid phosphatase activity, whereas the process of regeneration is characterized by an increase of NADH tetrazolium reductase, cytochrome oxidase, glutamate dehydrogenase and lactate dehydrogenase activities. The possible functional implications of these enzymes in the regenerative phenomena are discussed.  相似文献   

10.
Changes in number and distribution of retinal ganglion cells were studied after optic nerve crush in zebrafish (Brachydanio rerio) with retinal wholemount. There were approximately 40,000 to 56,000 cells in the retinal ganglion cell layer. The density of ganglion cells was divided into six classes and the area of highest cell density (central area) was located at the temporal area to the optic disc in normal fish. At the early regeneration stages after optic nerve crush, the percentage of lost cells increased gradually. Cell density had fallen first in the central area. At the late regeneration stages, there was an approximately 20% loss of ganglion cells during optic nerve regeneration. The results suggest that the loss of cells may undergo apoptosis rather than necrosis. A wave of cell loss started in the central area and spread progressively further into periphery. The reason caused these changes may be due to temporal interruption of optic nerve function, recovery from crush and the ability to quickly regenerate in optic nerve of the fish.  相似文献   

11.
Spermidine acts as an endogenous free radical scavenger and inhibits the action of reactive oxygen species. In this study, we examined the effects of spermidine on retinal ganglion cell (RGC) death in a mouse model of optic nerve injury (ONI). Daily ingestion of spermidine reduced RGC death following ONI and sequential in vivo retinal imaging revealed that spermidine effectively prevented retinal degeneration. Apoptosis signal-regulating kinase-1 (ASK1) is an evolutionarily conserved mitogen-activated protein kinase kinase kinase and has an important role in ONI-induced RGC apoptosis. We demonstrated that spermidine suppresses ONI-induced activation of the ASK1-p38 mitogen-activated protein kinase pathway. Moreover, production of chemokines important for microglia recruitment was decreased with spermidine treatment and, consequently, accumulation of retinal microglia is reduced. In addition, the ONI-induced expression of inducible nitric oxide synthase in the retina was inhibited with spermidine treatment, particularly in microglia. Furthermore, daily spermidine intake enhanced optic nerve regeneration in vivo. Our findings indicate that spermidine stimulates neuroprotection as well as neuroregeneration, and may be useful for treatment of various neurodegenerative diseases including glaucoma.Traumatic optic neuropathy is a common clinical problem that occurs in 0.5–5% of patients with closed head injury.1 A damage to the optic nerve causes shear stress and induces secondary swelling within the optic canal, accompanied by subsequent RGC loss and optic nerve atrophy.2 Although no large natural history or randomized controlled trial has been published, neither corticosteroid therapy nor optic canal decompression surgery is considered as standard treatments for patients with traumatic optic neuropathy,3 and there is a lack of effective treatment at present. Research into finding therapeutic targets for treatment of traumatic optic neuropathy indicated that neuroprotection and axon regeneration may be effective strategies and studies using an optic nerve injury (ONI) model in rodents have provided useful information. For example, neurotrophins, such as brain-derived neurotrophic factor and ciliary neurotrophic factor, protect retinal ganglion cells (RGCs) and promote axon regeneration in an ONI model.4, 5, 6 In addition, inhibition of neuroinflammatory events such as upregulation of tumor necrosis factor (TNF)-α and nitric oxide synthase (NOS) may be effective for RGC protection following ONI.7 The ONI model mimics some aspects of glaucoma, including RGC death induced by excitotoxicity and oxidative stress, and therefore it is also a useful animal model for glaucoma.Glaucoma is one of the leading causes of vision loss in the world and it is estimated that this condition will affect more than 80 million individuals worldwide by 2020, with at least 6–8 million individuals becoming bilaterally blind.8 Glaucoma is characterized by progressive degeneration of RGCs and their axons, which are usually associated with elevated intraocular pressure, but there is a subset of glaucoma termed normal tension glaucoma (NTG) that presents with statistically normal intraocular pressure. There are several animal models of glaucoma, including DBA/2J mice,9 and inducible models such as cauterization of episcleral veins.10, 11, 12 In addition, we previously reported that loss of glutamate transporters (EAAC1 or GLAST) in mice leads to RGC degeneration that is similar to NTG13 and these animal models have been useful in examining potential therapeutic targets.14, 15, 16Spermidine is naturally and almost exclusively accumulated in glial cells in the brain and retina.17, 18 It acts as an endogenous free radical scavenger and inhibits the action of reactive oxygen species. Indeed, it has been reported that spermidine has key roles in mediating protection against oxidative damage caused by hydrogen peroxide in cultured mouse fibroblasts19 and administration of spermidine extended the lifespan of yeast, flies, worms and human immune cells by upregulating the lysosomal/vacuolar degradation pathway, referred to as autophagy, which leads to enhanced resistance to oxidative stress and decreased cell death.20 Previously, we reported that oral administration of spermidine ameliorates severity of experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis, by suppression of oxidative stress,21 suggesting that spermidine may also be effective in protecting RGCs from increased oxidative stress associated with various pathogenic conditions in the eye including traumatic optic neuropathy and glaucoma.In this study, we examined the effects of daily spermidine intake on ONI-induced retinal degeneration. We monitored changes in retinal morphology over a course of 2 weeks following ONI, using optical coherence tomography (OCT), which permits noninvasive, longitudinal and quantitative assessment of retinal structures in living animals. We also explored possible mechanisms associated with spermidine-mediated neuroprotection.  相似文献   

12.
13.
GTP-binding protein Gem, a member protein of the Ras superfamily, can regulate actin cytoskeleton reorganization mediated by Rho-associated coiled-coil-containing protein kinase (ROCK). One attractive activity of the ROCK is playing a potential role in physiological and pathological process in retinal ganglion cells (RGCs) apoptosis. However, the function of Gem in retina is still with limited understanding. To investigate whether Gem is involved in optic nerve injury, we performed an optic nerve crush (ONC) model in adult rats. Western blot analysis indicated that Gem was significantly increased in the retina at the 3rd day after ONC. Meanwhile, double-immunofluorescent staining showed that Gem expression was mainly up-regulated in ganglion cell layer and co-localized with NeuN (a marker of RGCs). Additionally, the co-localizations of Gem/active-caspase-3 and Gem/TUNEL-positive cells were detected in RGCs. Furthermore, the expression of active-caspase-3 and TUNEL-positive cells was parallel with that of Gem. Finally, expression pattern of ROCK family (only ROCK2 but not ROCK1) was increased in the differentiated process, which was collected with the expression of GEM and active-caspase-3. Based on the present results, it is suggested that Gem might play a crucial role in RGCs apoptosis after ONC, which might be involved in ROCK pathway.  相似文献   

14.
In this study,the role of melanopsin-expressing retinal ganglion cells(mRGCs) in the glaucoma-induced depressive behavioral response pattern was investigated.The CFP-D2 transgenic glaucoma animal model from five age groups was used in this study.Immunohistochemical labeling,quantitative analysis of mRGC morphology,open field test(OFT),and statistical analysis were used.In comparison with C57 BL/6 mice,the age-matched CFP-D2 mice had significantly elevated intraocular pressure(IOP).We observed parallel morphological changes in the retina,including a reduction in the density of cyan fluorescent protein(CFP) expressing cells(cells mm 2 at 2 months of age,1309±26;14 months,878±30,P<0.001),mRGCs(2 months,48±3;14 months,19±4,P<0.001),Brn3b-expressing RGCs(2 months,1283±80;14 months,950±31,P<0.001),Brn-3b expressing mRGCs(5 months,50.17%±5.5%;14 months,12.61%±3.8%,P<0.001),and reduction in the dendritic field size of mRGCs(mm2 at 2 months,0.077±0.015;14 months,0.065±0.015,P<0.05).CFP-D2 mice had hyperactive locomotor activity patterns based on OFT findings of the total distance traveled,number of entries into the center,and time spent in the center of the testing apparatus.The glaucoma induced hyperactive response pattern could be associated with dysfunctional mRGCs,most likely Brn-3b-positive mRGCs in CFP-D2 mice.  相似文献   

15.
Damage to the optic nerve in mammals induces retrograde degeneration and apoptosis of the retinal ganglion cell (RGC) bodies. The mechanisms that mediate the response of the neuronal cells to the axonal injury are still unknown. We have previously shown that semaphorins, axon guidance molecules with repulsive cues, are capable of mediating apoptosis in cultured neuronal cells (Shirvan, A., Ziv, I., Fleminger, G., Shina, R., He, Z., Brudo, I., Melamed, E., and Brazilai, A. (1999) J. Neurochem. 73, 961-971). In this study, we examined the involvement of semaphorins in an in vivo experimental animal model of complete axotomy of the rat optic nerve. We demonstrate that a marked induction of type III semaphorin proteins takes place in ipsilateral retinas at early stages following axotomy, well before any morphological signs of RGC apoptosis can be detected. Time course analysis revealed that a peak of expression occurred after 2-3 days and then declined. A small conserved peptide derived from semaphorin 3A that was previously shown to induce neuronal death in culture was capable of inducing RGC loss upon its intravitreous injection into the rat eye. Moreover, we demonstrate a marked inhibition of RGC loss when axotomized eyes were co-treated by intravitreous injection of function-blocking antibodies against the semaphorin 3A-derived peptide. Marked neuronal protection from degeneration was also observed when the antibodies were applied 24 h post-injury. We therefore suggest that semaphorins are key proteins that modulate the cell fate of axotomized RGC. Neutralization of the semaphorin repulsive function may serve as a promising new approach for treatment of traumatic injury in the adult mammalian central nervous system or of ophthalmologic diseases such as glaucoma and ischemic optic neuropathy that induce apoptotic RGC death.  相似文献   

16.
Axonal degeneration of retinal ganglion cells (RGCs) causes blindness in glaucoma. Currently, there are no therapies that target axons to prevent them from degenerating. Activation of the BAX protein has been shown to be the determining step in the intrinsic apoptotic pathway that causes RGCs to die in glaucoma. A putative role for BAX in axonal degeneration is less well elucidated. BCLXL (BCL2L1) is the primary antagonist of BAX in RGCs. We developed a mCherry-BCLXL fusion protein, which prevented BAX recruitment and activation to the mitochondria in tissue culture cells exposed to staurosporine. This fusion protein was then packaged into adeno-associated virus serotype 2, which was used to transduce RGCs after intravitreal injection and force its overexpression. Transduced RGCs express mCherry-BCLXL throughout their somas and axons along the entire optic tract. In a model of acute optic nerve crush, the transgene prevented the recruitment of a GFP-BAX fusion protein to mitochondria and provided long-term somal protection up to 12 weeks post injury. To test the efficacy in glaucoma, DBA/2J mice were transduced at 5 months of age, just prior to the time they begin to exhibit ocular hypertension. Gene therapy with mCherry-BCLXL did not affect the longitudinal history of intraocular pressure elevation compared to naive mice but did robustly attenuate both RGC soma pathology and axonal degeneration in the optic nerve at both 10.5 and 12 months of age. BCLXL gene therapy is a promising candidate for glaucoma therapy.Subject terms: Cell death in the nervous system, Neurodegeneration  相似文献   

17.
Retinal ganglion cells (RGCs) are central nervous system (CNS) neurons that transmit visual information from the retina to the brain. Apoptotic RGC degeneration causes visual impairment that can be modeled by optic nerve crush. Neuronal apoptosis is also a salient feature of CNS trauma, ischemia (stroke), and diseases of the CNS such as Alzheimer's, Parkinson's, multiple sclerosis, and amyotrophic lateral sclerosis. Optic nerve crush induces the apoptotic cell death of ~ 70% of RGCs within the first 14 days after injury. This model is particularly attractive for studying adult neuron apoptosis because the time-course of RGC death is well established and axon regeneration within the myelinated optic nerve can be concurrently evaluated. Here, we performed a large scale iTRAQ proteomic study to identify and quantify proteins of the rat retina at 1, 3, 4, 7, 14, and 21 days after optic nerve crush. In total, 337 proteins were identified, and 110 were differentially regulated after injury. Of these, 58 proteins were upregulated (>1.3 ×), 46 were downregulated (<0.7 ×), and 6 showed both positive and negative regulation over 21 days, relative to normal retinas. Among the differentially expressed proteins, Thymosin-β4 showed an early upregulation at 3 days, the time-point that immediately precedes the induction of RGC apoptosis after injury. We examined the effect of exogenous Thymosin-β4 administration on RGC death after optic nerve injury. Intraocular injections of Thymosin-β4 significantly increased RGC survival by ~ 3-fold compared to controls and enhanced axon regeneration after crush, demonstrating therapeutic potential for CNS insults. Overall, our study identified numerous proteins that are differentially regulated at key time-points after optic nerve crush, and how the temporal profiles of their expression parallel RGC death. This data will aid in the future development of novel therapeutics to promote neuronal survival and regeneration in the adult CNS.  相似文献   

18.
Recent advances in mouse genomics have revealed considerable variation in the form of single-nucleotide polymorphisms (SNPs) among common inbred strains. This has made it possible to characterize closely related strains and to identify genes that differ; such genes may be causal for quantitative phenotypes. The mouse strains DBA/1J and DBA/2J differ by just 5.6% at the SNP level. These strains exhibit differences in a number of metabolic and lipid phenotypes, such as plasma levels of triglycerides (TGs) and HDL. A cross between these strains revealed multiple quantitative trait loci (QTLs) in 294 progeny. We identified significant TG QTLs on chromosomes (Chrs) 1, 2, 3, 4, 8, 9, 10, 11, 12, 13, 14, 16, and 19, and significant HDL QTLs on Chrs 3, 9, and 16. Some QTLs mapped to chromosomes with limited variability between the two strains, thus facilitating the identification of candidate genes. We suggest that Tshr is the QTL gene for Chr 12 TG and HDL levels and that Ihh may account for the TG QTL on Chr 1. This cross highlights the advantage of crossing closely related strains for subsequent identification of QTL genes.  相似文献   

19.
Imaging has gained a key role in modern glaucoma management. Traditionally, interest was directed toward the appearance of the optic nerve head and the retinal nerve fiber layer. With the improvement of the resolution of optical coherence tomography, the ganglion cell complex has also become routinely accessible in the clinic. Further advances have been made in understanding the structure-function relationship in glaucoma. Nevertheless, direct imaging of the retinal ganglion cells in glaucoma would be advantageous. With the currently used techniques, this goal cannot be achieved, because the transversal resolution is limited by aberrations of the eye. The use of adaptive optics has significantly improved transversal resolution, and the imaging of several cell types including cones and astrocytes has become possible. Imaging of retinal ganglion cells, however, still remains a problem, because of the transparency of these cells. However, the visualization of retinal ganglion cells and their dendrites has been achieved in animal models. Furthermore, attempts have been made to visualize the apoptosis of retinal ganglion cells in vivo. Implementation of these techniques in clinical practice will probably improve glaucoma care and facilitate the development of neuroprotective strategies.  相似文献   

20.
Cell-based therapies are increasingly recognized as a potential strategy to treat retinal neurodegenerative disease. Their administration, however, is normally indirect and complex, often with an inability to assess in real time their effects on cell death and their migration/integration into the host retina. In the present study, using a partial optic nerve transection (pONT) rat model, we describe a new method of Schwann cell (SC) delivery (direct application to injured optic nerve sheath, SC/DONS), which was compared with intravitreal SC delivery (SC/IVT). Both SC/DONS and SC/IVT were able to be assessed in vivo using imaging to visualize retinal ganglion cell (RGC) apoptosis and SC retinal integration. RGC death in the pONT model was best fitted to the one-phase exponential decay model. Although both SC/DONS and SC/IVT altered the temporal course of RGC degeneration in pONT, SC/DONS resulted in delayed but long-lasting effects on RGC protection, compared with SC/IVT treatment. In addition, their effects on primary and secondary degeneration, and axonal regeneration, were also investigated, by histology, whole retinal counting, and modelling of RGC loss. SC/DONS was found to significantly reduce RGC apoptosis in vivo and significantly increase RGC survival by targeting secondary rather than primary degeneration. Both SC/DONS and SC/IVT were found to promote RGC axonal regrowth after optic nerve injury, with evidence of GAP-43 expression in RGC somas and axons. SC/DONS may have the potential in the treatment of optic neuropathies, such as glaucoma. We show that SC transplantation can be monitored in real time and that the protective effects of SCs are associated with targeting secondary degeneration, with implications for translating cell-based therapies to the clinic.In the central (CNS) and peripheral (PNS) nervous systems, injury from initial lesions can lead to widespread damage to neurons beyond the primary injury site; a phenomenon known as ‘secondary degeneration''. Studies in spinal cord injury have revealed secondary rather than primary degeneration to be the major contributor to neuronal death and functional impairment, and it is increasingly recognized as a therapeutic target.1,2 Secondary degeneration also occurs in optic neuropathies, including glaucoma, ischaemic optic neuropathy, and Leber''s hereditary optic neuropathy.3, 4, 5 Retinal neuronal loss in these conditions is reported to occur long after the initial insult,6 implying that secondary mechanisms may have an important role in optic neuropathic damage and that targeting of secondary neuronal loss may represent a novel therapeutic strategy.Partial optic nerve transection (pONT) represents a reliable and reproducible model for studying secondary degeneration, in which a primary lesion is only made to dorsal axons and leaves those in ventral optic nerve (ON) intact but vulnerable to secondary degeneration.4,7 Secondary degeneration is thought to be initiated by a cascade of reactive metabolic events, including glutamate excitotoxicity, Ca2+ overload, excess free radical formation, oxidative stress, mitochondrial dysfunction, and increased proteoglycan expression, leading to cell death.7, 8, 9, 10, 11, 12, 13, 14 Activated astrocytes are reported to be a major contributor to spreading and acceleration of secondary degeneration.8,9As in most CNS pathways, the mature ON possesses only a limited ability to repair itself after injury, resulting in permanent vision loss due to the death of retinal ganglion cells (RGCs), the retinal output neurons that transmit visual information to the brain.15 Compared with the CNS, the PNS has a remarkable ability to regrow after injury, a process in which Schwann cells (SCs) are thought to have a key role.16,17SCs are the principal glia of the PNS and support normal neuronal function.18,19 Upon axonal injury, SCs are reported to shed their myelin sheaths and de-differentiate into progenitor stem cells, which are capable of replacing damaged tissue and providing a permissive environment for neuronal survival and axonal regrowth.18,19 SCs are believed to achieve this through releasing neurotrophic factors and producing cell adhesion molecules and extracellular matrix components.20 The neuroprotective and regenerative mechanisms between SCs and neurons are thought to operate on a local basis via adhesion molecules, allowing contact-mediated signalling between cells,16,17,20,21 and extracellular free ligands, facilitating specific binding to the receptors in the target neurons.16,17,20 However, a novel regulatory mechanism has emerged, representing a more efficient and advanced communication machinery, that is, vesicular transfer between SCs and axons.16 We have recently demonstrated that the highly efficient response of SCs to PN injury is triggered by Ephrin-B/EphB2 signalling in fibroblasts, which guide SC sorting and migration during nerve repair.21Due to the regenerative ability of SCs in PNS repair, transplantation of SCs to the injured ON has been previously attempted.22, 23, 24, 25, 26, 27, 28 To date, however, the protective effects of SCs on retinal neurons have been only assessed after either intravitreal administration or suturing artificial SC grafts onto transected ON, using postmortem histological observations, with incomplete delineation of the mechanisms involved.22, 23, 24, 25, 26, 27, 28Here we use a pONT model to investigate a new method of SC delivery (direct application to injured ON sheath, SC/DONS), using in vivo imaging and histological techniques, and compare its effects on RGC apoptosis and loss to intravitreal SC delivery (SC/IVT). Furthermore, we analyse whether these actions target primary or secondary degeneration, to determine their potential in the treatment of optic neuropathy.  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