Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 176
Filter
1.
Exp Eye Res ; 244: 109929, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38750783

ABSTRACT

Optic nerve injuries are severely disrupt the structural and functional integrity of the retina, often leading to visual impairment or blindness. Despite the profound impact of these injuries, the molecular mechanisms involved remain poorly understood. In this study, we performed a comprehensive whole-transcriptome analysis of mouse retina samples after optic nerve crush (ONC) to elucidate changes in gene expression and regulatory networks. Transcriptome analysis revealed a variety of molecular alterations, including 256 mRNAs, 530 lncRNAs, and 37 miRNAs, associated with metabolic, inflammatory, signaling, and biosynthetic pathways in the injured retina. The integrated analysis of co-expression and protein-protein interactions identified an active interconnected module comprising 5 co-expressed proteins (Fga, Serpina1a, Hpd, Slc38a4, and Ahsg) associated with the complement and coagulation cascades. Finally, 5 mRNAs (Fga, Serpinala, Hpd, Slc38a4, and Ahsg), 2 miRNAs (miR-671-5p and miR-3057-5p), and 6 lncRNAs (MSTRG. 1830.1, Gm10814, A530013C23Rik, Gm40634, MSTRG.9514.1, A330023F24Rik) were identified by qPCR in the injured retina, and some of them were validated as critical components of a ceRNA network active in 661W and HEK293T cells through dual-luciferase reporter assays. In conclusion, our study provides comprehensive insight into the complex and dynamic biological mechanisms involved in retinal injury responses and highlights promising potential targets to enhance neuroprotection and restore vision.


Subject(s)
Gene Expression Profiling , Gene Regulatory Networks , Mice, Inbred C57BL , Optic Nerve Injuries , RNA, Messenger , Retina , Animals , Mice , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/genetics , Retina/metabolism , RNA, Messenger/genetics , Disease Models, Animal , Transcriptome , MicroRNAs/genetics , Gene Expression Regulation/physiology , Male , Humans , RNA, Long Noncoding/genetics
2.
Mol Neurobiol ; 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38639863

ABSTRACT

Retinal ganglion cells (RGCs), neurons transmitting visual information via the optic nerve, fail to regenerate their axons after injury. The progressive loss of RGC function underlies the pathophysiology of glaucoma and other optic neuropathies, often leading to irreversible blindness. Therefore, there is an urgent need to identify the regulators of RGC survival and the regenerative program. In this study, we investigated the role of the family of transcription factors known as nuclear factor of activated T cells (NFAT), which are expressed in the retina; however, their role in RGC survival after injury is unknown. Using the optic nerve crush (ONC) model, widely employed to study optic neuropathies and central nervous system axon injury, we found that NFATc4 is specifically but transiently up-regulated in response to mechanical injury. In the injured retina, NFATc4 immunolocalized primarily to the ganglionic cell layer. Utilizing NFATc4-/- and NFATc3-/- mice, we demonstrated that NFATc4, but not NFATc3, knockout increased RGC survival, improved retina function, and delayed axonal degeneration. Microarray screening data, along with decreased immunostaining of cleaved caspase-3, revealed that NFATc4 knockout was protective against ONC-induced degeneration by suppressing pro-apoptotic signaling. Finally, we used lentiviral-mediated NFATc4 delivery to the retina of NFATc4-/- mice and reversed the pro-survival effect of NFATc4 knockout, conclusively linking the enhanced survival of injured RGCs to NFATc4-dependent mechanisms. In summary, this study is the first to demonstrate that NFATc4 knockout may confer transient RGC neuroprotection and decelerate axonal degeneration after injury, providing a potent therapeutic strategy for optic neuropathies.

3.
Heliyon ; 10(8): e29428, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38638966

ABSTRACT

Activated astrocytes are a primary source of inflammatory factors following traumatic optic neuropathy (TON). Accumulation of inflammatory factors in this context leads to increased axonal damage and loss of retinal ganglion cells (RGCs). Therefore, in the present study, we explored the role of the astrocyte G protein-coupled estrogen receptor (GPER) in regulating inflammatory factors following optic nerve crush (ONC), and analyzed its potential regulatory mechanisms. Overall, our results showed that GPER was abundantly expressed in the optic nerve, and co-localized with glial fibrillary acidic proteins (GFAP). Exogenous administration of G-1 led to a significant reduction in astrocyte activation and expression of inflammation-related factors (including IL-1ß, TNF-α, NFκB, and p-NFκB). Additionally, it dramatically increased the survival of RGCs. In contrast, astrocytes were activated to a greater extent by exogenous G15 administration; however, RGCs survival was significantly reduced. In vitro, GPER activation significantly reduced astrocyte activation and the release of inflammation-related factors. In conclusion, activation of astrocyte GPER significantly reduced ONC inflammation levels, and should be explored as a potential target pathway for protecting the optic nerve and RGCs after TON.

4.
eNeuro ; 11(3)2024 Mar.
Article in English | MEDLINE | ID: mdl-38548335

ABSTRACT

Neuroprotection after injury or in neurodegenerative disease remains a major goal for basic and translational neuroscience. Retinal ganglion cells (RGCs), the projection neurons of the eye, degenerate in optic neuropathies after axon injury, and there are no clinical therapies to prevent their loss or restore their connectivity to targets in the brain. Here we demonstrate a profound neuroprotective effect of the exogenous expression of various Ca2+/calmodulin-dependent protein kinase II (CaMKII) isoforms in mice. A dramatic increase in RGC survival following the optic nerve trauma was elicited by the expression of constitutively active variants of multiple CaMKII isoforms in RGCs using adeno-associated viral (AAV) vectors across a 100-fold range of AAV dosing in vivo. Despite this neuroprotection, however, short-distance RGC axon sprouting was suppressed by CaMKII, and long-distance axon regeneration elicited by several pro-axon growth treatments was likewise inhibited even as CaMKII further enhanced RGC survival. Notably, in a dose-escalation study, AAV-expressed CaMKII was more potent for axon growth suppression than the promotion of survival. That diffuse overexpression of constitutively active CaMKII strongly promotes RGC survival after axon injury may be clinically valuable for neuroprotection per se. However, the associated strong suppression of the optic nerve axon regeneration demonstrates the need for understanding the intracellular domain- and target-specific CaMKII activities to the development of CaMKII signaling pathway-directed strategies for the treatment of optic neuropathies.


Subject(s)
Neurodegenerative Diseases , Optic Nerve Diseases , Optic Nerve Injuries , Mice , Animals , Retinal Ganglion Cells/metabolism , Optic Nerve Injuries/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Axons/metabolism , Neurodegenerative Diseases/metabolism , Nerve Regeneration/physiology , Optic Nerve Diseases/metabolism , Protein Isoforms/metabolism , Cell Survival/physiology
5.
Exp Neurol ; 375: 114741, 2024 May.
Article in English | MEDLINE | ID: mdl-38395216

ABSTRACT

Nuclear factor erythroid 2 like (Nfe2l) gene family members 1-3 mediate cellular response to oxidative stress, including in the central nervous system (CNS). However, neuronal functions of Nfe2l3 are unknown. Here, we comparatively evaluated expression of Nfe2l1, Nfe2l2, and Nfe2l3 in singe cell RNA-seq (scRNA-seq)-profiled cortical and retinal ganglion cell (RGC) CNS projection neurons, investigated whether Nfe2l3 regulates neuroprotection and axon regeneration after CNS injury in vivo, and characterized a gene network associated with Nfe2l3 in neurons. We showed that, Nfe2l3 expression transiently peaks in developing immature cortical and RGC projection neurons, but is nearly abolished in adult neurons and is not upregulated after injury. Furthermore, within the retina, Nfe2l3 is enriched in RGCs, primarily neonatally, and not upregulated in injured RGCs, whereas Nfe2l1 and Nfe2l2 are expressed robustly in other retinal cell types as well and are upregulated in injured RGCs. We also found that, expressing Nfe2l3 in injured RGCs through localized intralocular viral vector delivery promotes neuroprotection and long-distance axon regeneration after optic nerve injury in vivo. Moreover, Nfe2l3 provided a similar extent of neuroprotection and axon regeneration as viral vector-targeting of Pten and Klf9, which are prominent regulators of neuroprotection and long-distance axon regeneration. Finally, we bioinformatically characterized a gene network associated with Nfe2l3 in neurons, which predicted the association of Nfe2l3 with established mechanisms of neuroprotection and axon regeneration. Thus, Nfe2l3 is a novel neuroprotection and axon regeneration-promoting factor with a therapeutic potential for treating CNS injury and disease.


Subject(s)
Axons , Optic Nerve Injuries , Humans , Axons/physiology , Neuroprotection , Nerve Regeneration/physiology , Retinal Ganglion Cells/metabolism , Retina/metabolism , Optic Nerve Injuries/metabolism
6.
Exp Eye Res ; 239: 109787, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38211683

ABSTRACT

Retinal ganglion cell (RGC) death and axonal loss cause irreversible vision loss upon optic nerve (ON) injury. We have independently demonstrated that mesenchymal stem cells (MSCs) and green tea extract (GTE) promote RGC survival and axonal regeneration in rats with ON injury. Here we aimed to evaluate the combined treatment effect of human bone marrow-derived MSCs (hBM-MSCs) and GTE on RGC survival and axonal regeneration after ON injury. Combined treatment of hBM-MSCs and GTE promoted RGC survival and neurite outgrowth/axonal regeneration in ex vivo retinal explant culture and in rats after ON injury. GTE increased Stat3 activation in the retina after combined treatment, and enhanced brain-derived neurotrophic factor secretion from hBM-MSCs. Treatment of 10 µg/mL GTE would not induce hBM-MSC apoptosis, but inhibited their proliferation, migration, and adipogenic and osteogenic differentiation in vitro with reducing matrix metalloproteinase secretions. In summary, this study revealed that GTE can enhance RGC protective effect of hBM-MSCs, suggesting that stem cell priming could be a prospective strategy enhancing the properties of stem cells for ON injury treatment.


Subject(s)
Mesenchymal Stem Cells , Optic Nerve Injuries , Rats , Humans , Animals , Optic Nerve Injuries/therapy , Optic Nerve Injuries/metabolism , Retinal Ganglion Cells/metabolism , Osteogenesis , Tea/metabolism , Nerve Regeneration/physiology , Cell Survival/physiology , Axons/metabolism
7.
Craniomaxillofac Trauma Reconstr ; 16(4): 306-316, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38047144

ABSTRACT

Study Design: A systematic review and meta-analysis. Objective: Treatment of traumatic optic neuropathy (TON) has been a subject of debate for many decades due to the scarcity of evidence-based treatment protocols. This review compares surgical decompression (SD) and steroid therapy (ST) as treatment approaches in TON patients. Methods: A PRISMA-guided systematic review using PubMed, Embase, Ovid and Scopus databases was performed till the last search date of July 31st 2021. The outcome of interest was an improvement in visual acuity. A meta-analysis of the odds ratio was performed using a random-effect model and sub-group analysis based upon criteria for assessment of improvement in visual acuity. Results: Sixteen studies (including 1046 patients) were included in the review. The review could identify 590 patients treated with SD and 456 treated with ST. In addition, there was a second cohort of patients presenting with NLP (no light perception). A meta-analysis with a sub-group analysis revealed that there was statistically no significant difference between the two treatment approaches in terms of improvement in VA. Conclusions: There is no difference in treatment results of SD or ST for TON. Several treatment protocols and different criteria for assessing visual acuity led to difficulty in generating evidence for selecting the correct treatment approach.

8.
Fa Yi Xue Za Zhi ; 39(4): 350-359, 2023 Aug 25.
Article in English, Chinese | MEDLINE | ID: mdl-37859473

ABSTRACT

OBJECTIVES: To investigate the characteristics and objective assessment method of visual field defects caused by optic chiasm and its posterior visual pathway injury. METHODS: Typical cases of visual field defects caused by injuries to the optic chiasm, optic tracts, optic radiations, and visual cortex were selected. Visual field examinations, visual evoked potential (VEP) and multifocal visual evolved potential (mfVEP) measurements, craniocerebral CT/MRI, and retinal optical coherence tomography (OCT) were performed, respectively, and the aforementioned visual electrophysiological and neuroimaging indicators were analyzed comprehensively. RESULTS: The electrophysiological manifestations of visual field defects caused by optic chiasm injuries were bitemporal hemianopsia mfVEP abnormalities. The visual field defects caused by optic tract, optic radiation, and visual cortex injuries were all manifested homonymous hemianopsia mfVEP abnormalities contralateral to the lesion. Mild relative afferent pupil disorder (RAPD) and characteristic optic nerve atrophy were observed in hemianopsia patients with optic tract injuries, but not in patients with optic radiation or visual cortex injuries. Neuroimaging could provide morphological evidence of damages to the optic chiasm and its posterior visual pathway. CONCLUSIONS: Visual field defects caused by optic chiasm, optic tract, optic radiation, and visual cortex injuries have their respective characteristics. The combined application of mfVEP and static visual field measurements, in combination with neuroimaging, can maximize the assessment of the location and degree of visual pathway damage, providing an effective scheme for the identification of such injuries.


Subject(s)
Brain Injuries, Traumatic , Optic Nerve Injuries , Humans , Optic Chiasm/diagnostic imaging , Optic Chiasm/pathology , Visual Pathways/diagnostic imaging , Visual Pathways/pathology , Visual Fields , Evoked Potentials, Visual , Random Amplified Polymorphic DNA Technique , Hemianopsia/etiology , Hemianopsia/complications , Vision Disorders/diagnosis , Vision Disorders/etiology , Vision Disorders/pathology , Optic Nerve Injuries/diagnostic imaging , Brain Injuries, Traumatic/diagnosis , Brain Injuries, Traumatic/diagnostic imaging
9.
Elife ; 122023 10 17.
Article in English | MEDLINE | ID: mdl-37846146

ABSTRACT

Injured axons in the central nervous system (CNS) usually fail to regenerate, causing permanent disabilities. However, the knockdown of Pten knockout or treatment of neurons with hyper-IL-6 (hIL-6) transforms neurons into a regenerative state, allowing them to regenerate axons in the injured optic nerve and spinal cord. Transneuronal delivery of hIL-6 to the injured brain stem neurons enables functional recovery after severe spinal cord injury. Here we demonstrate that the beneficial hIL-6 and Pten knockout effects on axon growth are limited by the induction of tubulin detyrosination in axonal growth cones. Hence, cotreatment with parthenolide, a compound blocking microtubule detyrosination, synergistically accelerates neurite growth of cultured murine CNS neurons and primary RGCs isolated from adult human eyes. Systemic application of the prodrug dimethylamino-parthenolide (DMAPT) facilitates axon regeneration in the injured optic nerve and spinal cord. Moreover, combinatorial treatment further improves hIL-6-induced axon regeneration and locomotor recovery after severe SCI. Thus, DMAPT facilitates functional CNS regeneration and reduces the limiting effects of pro-regenerative treatments, making it a promising drug candidate for treating CNS injuries.


Subject(s)
Axons , Spinal Cord Injuries , Mice , Animals , Humans , Axons/physiology , Nerve Regeneration , Neurons/physiology , Spinal Cord Injuries/drug therapy , Microtubules
10.
Exp Neurol ; 368: 114510, 2023 10.
Article in English | MEDLINE | ID: mdl-37633482

ABSTRACT

Ribosomal proteins are involved in neurodevelopment and central nervous system (CNS) disease and injury. However, the roles of specific ribosomal protein subunits in developmental axon growth, and their potential as therapeutic targets for treating CNS injuries, are still poorly understood. Here, we show that ribosomal protein large (Rpl) and small (Rps) subunit genes are substantially (56-fold) enriched amongst the genes, which are downregulated during maturation of retinal ganglion cell (RGC) CNS projection neurons. We also show that Rpl and Rps subunits are highly co-regulated in RGCs, and partially re-upregulated after optic nerve crush (ONC). Because developmental downregulation of ribosomal proteins coincides with developmental decline in neuronal intrinsic axon growth capacity, we hypothesized that Rpl/Rps incomplete re-upregulation after injury may be a part of the cellular response which attempts to reactivate intrinsic axon growth mechanisms. We found that experimentally upregulating Rpl7 and Rpl7A promoted axon regeneration after ONC in vivo. Finally, we characterized gene networks associated with Rpl/Rps, and showed that Rpl7 and Rpl7A belong to the cluster of genes, which are shared between translational and neurodevelopmental biological processes (based on gene-ontology) that are co-downregulated in maturing RGCs during the decline in intrinsic axon growth capacity.


Subject(s)
Axons , Nerve Regeneration , Up-Regulation , Nerve Regeneration/genetics , Transcriptional Activation , Ribosomal Proteins/genetics
11.
Methods Mol Biol ; 2708: 25-31, 2023.
Article in English | MEDLINE | ID: mdl-37558956

ABSTRACT

Organotypic retinal explants are routinely used as alternatives to in vitro cell culture and to replace the use of animals in modelling retinal neurodegenerative diseases. Retinal explants fill the gap between in vivo which are expensive, time consuming, and complex due to inaccessibility of target tissues. However, organotypic retinal explants are less expensive and rapid and retinal cell types in the explant maintain their morphologic interactions with other cells in the retina. Therefore, retinal explants have high potential to be used as tools to assess the pharmacological and other therapies prior to in vivo validation, reducing the use of live animals.


Subject(s)
Retina , Animals , Organ Culture Techniques , Retina/metabolism
12.
Exp Ther Med ; 26(2): 381, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37456161

ABSTRACT

Optic nerve injury is a type of neurodegenerative disease. Physcion is an anthraquinone that exerts a protective role against various diseases. However, its function in regulating optic nerve injury remains largely unknown. An in vitro model of optic nerve injury was established in HAPI cells treated with IFN-ß. Functional assays were used to detect HAPI cell viability and apoptosis. The levels of inflammation and the expression levels of oxidative stress-related genes were measured in HAPI cells. In addition, western blot analysis was used to detect the expression levels of Janus kinase 2 (JAK2)/STAT3-linked genes in HAPI cells. Treatment of the cells with physcion prevented cells against IFN-ß-induced neuronal injury. Physcion restrained IFN-ß-induced inflammatory response and oxidative stress in HAPI cells. In addition, it improved IFN-ß-induced injury in HAPI cells by suppressing the JAK2/STAT3 pathway. In conclusion, the present study revealed that physcion improved optic nerve injury in vitro by inhibiting the JAK2/STAT3 pathway. Physcion may be a promising therapeutic target for the treatment of this disease.

13.
Neural Regen Res ; 18(12): 2773-2780, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37449644

ABSTRACT

Vision depends on accurate signal conduction from the retina to the brain through the optic nerve, an important part of the central nervous system that consists of bundles of axons originating from retinal ganglion cells. The mammalian optic nerve, an important part of the central nervous system, cannot regenerate once it is injured, leading to permanent vision loss. To date, there is no clinical treatment that can regenerate the optic nerve and restore vision. Our previous study found that the mobile zinc (Zn2+) level increased rapidly after optic nerve injury in the retina, specifically in the vesicles of the inner plexiform layer. Furthermore, chelating Zn2+ significantly promoted axonal regeneration with a long-term effect. In this study, we conditionally knocked out zinc transporter 3 (ZnT3) in amacrine cells or retinal ganglion cells to construct two transgenic mouse lines (VGATCreZnT3fl/fl and VGLUT2CreZnT3fl/fl, respectively). We obtained direct evidence that the rapidly increased mobile Zn2+ in response to injury was from amacrine cells. We also found that selective deletion of ZnT3 in amacrine cells promoted retinal ganglion cell survival and axonal regeneration after optic nerve crush injury, improved retinal ganglion cell function, and promoted vision recovery. Sequencing analysis of reginal ganglion cells revealed that inhibiting the release of presynaptic Zn2+ affected the transcription of key genes related to the survival of retinal ganglion cells in postsynaptic neurons, regulated the synaptic connection between amacrine cells and retinal ganglion cells, and affected the fate of retinal ganglion cells. These results suggest that amacrine cells release Zn2+ to trigger transcriptomic changes related to neuronal growth and survival in reginal ganglion cells, thereby influencing the synaptic plasticity of retinal networks. These results make the theory of zinc-dependent retinal ganglion cell death more accurate and complete and provide new insights into the complex interactions between retinal cell networks.

14.
Clin Exp Ophthalmol ; 51(6): 627-641, 2023 08.
Article in English | MEDLINE | ID: mdl-37317890

ABSTRACT

The retinal ganglion cells (RGCs) are the sole output neurons that connect information from the retina to the brain. Optic neuropathies such as glaucoma, trauma, inflammation, ischemia and hereditary optic neuropathy can cause RGC loss and axon damage, and lead to partial or total loss of vision, which is an irreversible process in mammals. The accurate diagnoses of optic neuropathies are crucial for timely treatments to prevent irrevocable RGCs loss. After severe ON damage in optic neuropathies, promoting RGC axon regeneration is vital for restoring vision. Clearance of neuronal debris, decreased intrinsic growth capacity, and the presence of inhibitory factors have been shown to contribute to the failure of post-traumatic CNS regeneration. Here, we review the current understanding of manifestations and treatments of various common optic neuropathies. We also summarise the current known mechanisms of RGC survival and axon regeneration in mammals, including specific intrinsic signalling pathways, key transcription factors, reprogramming genes, inflammation-related regeneration factors, stem cell therapy, and combination therapies. Significant differences in RGC subtypes in survival and regenerative capacity after injury have also been found. Finally, we highlight the developmental states and non-mammalian species that are capable of regenerating RGC axons after injury, and cellular state reprogramming for neural repair.


Subject(s)
Optic Nerve Diseases , Optic Nerve Injuries , Humans , Animals , Axons , Optic Nerve Injuries/therapy , Optic Nerve Injuries/metabolism , Nerve Regeneration/physiology , Mammals
15.
Front Neurosci ; 17: 1158030, 2023.
Article in English | MEDLINE | ID: mdl-37090805

ABSTRACT

Gene therapy has become an essential treatment for optic nerve injury (ONI) in recent years, and great strides have been made using animal models. ONI, which is characterized by the loss of retinal ganglion cells (RGCs) and axons, can induce abnormalities in the pupil light reflex, visual field defects, and even vision loss. The eye is a natural organ to target with gene therapy because of its high accessibility and certain immune privilege. As such, numerous gene therapy trials are underway for treating eye diseases such as glaucoma. The aim of this review was to cover research progress made in gene therapy for ONI. Specifically, we focus on the potential of gene therapy to prevent the progression of neurodegenerative diseases and protect both RGCs and axons. We cover the basic information of gene therapy, including the classification of gene therapy, especially focusing on genome editing therapy, and then we introduce common editing tools and vector tools such as Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) -Cas9 and adeno-associated virus (AAV). We also summarize the progress made on understanding the roles of brain derived neurotrophic factor (BDNF), ciliary neurotrophic factor (CNTF), phosphatase-tensin homolog (PTEN), suppressor of cytokine signal transduction 3 (SOCS3), histone acetyltransferases (HATs), and other important molecules in optic nerve protection. However, gene therapy still has many challenges, such as misalignment and mutations, immunogenicity of AAV, time it takes and economic cost involved, which means that these issues need to be addressed before clinical trials can be considered.

16.
Brain Res ; 1809: 148368, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37059258

ABSTRACT

Collapsin response mediator proteins (Crmps) play roles in neuronal development and axon growth. However, neuronal-specific roles of Crmp1, Crmp4, and Crmp5 in regeneration of injured central nervous system (CNS) axons in vivo are unclear. Here, we analyzed developmental and subtype-specific expression of Crmp genes in retinal ganglion cells (RGCs), tested whether overexpressing Crmp1, Crmp4, or Crmp5 in RGCs through localized intralocular AAV2 delivery promotes axon regeneration after optic nerve injury in vivo, and characterized developmental co-regulation of gene-concept networks associated with Crmps. We found that all Crmp genes are developmentally downregulated in RGCs during maturation. However, while Crmp1, Crmp2, and Crmp4 were expressed to a varying degree in most RGC subtypes, Crmp3 and Crmp5 were expressed only in a small subset of RGC subtypes. We then found that after optic nerve injury, Crmp1, Crmp4, and Crmp5 promote RGC axon regeneration to varying extents, with Crmp4 promoting the most axon regeneration and also localizing to axons. We also found that Crmp1 and Crmp4, but not Crmp5, promote RGC survival. Finally, we found that Crmp1, Crmp2, Crmp4, and Crmp5's ability to promote axon regeneration is associated with neurodevelopmental mechanisms, which control RGC's intrinsic axon growth capacity.


Subject(s)
Optic Nerve Injuries , Retinal Ganglion Cells , Humans , Retinal Ganglion Cells/metabolism , Axons/metabolism , Optic Nerve Injuries/metabolism , Nerve Regeneration/physiology , Gene Expression , Cell Survival
17.
Methods Mol Biol ; 2636: 205-219, 2023.
Article in English | MEDLINE | ID: mdl-36881302

ABSTRACT

Many human optic neuropathies lead to crippling conditions resulting in partial or complete loss of vision. While the retina is made up of several different cell types, retinal ganglion cells (RGCs) are the only cell type connecting the eye to the brain. Optic nerve crush injuries, wherein RGC axons are damaged without severing the optic nerve sheath, can serve as a model for traumatic optical neuropathies as well as some progressive neuropathies such as glaucoma. In this chapter, we describe two different surgical methods for establishing an optic nerve crush (ONC) injury in the postmetamorphic frog, Xenopus laevis. Why use the frog as an animal model? Mammals lose the ability to regenerate damaged CNS neurons, but amphibians and fish retain the ability to regenerate new RGC bodies and regrow RGC axons following an injury. In addition to presenting two different surgical ONC injury methods, we highlight their advantages and disadvantages and discuss the distinctive characteristics of Xenopus laevis as an animal model for studying CNS regeneration.


Subject(s)
Crush Injuries , Peripheral Nerve Injuries , Animals , Humans , Xenopus laevis , Retina/surgery , Anura , Optic Nerve , Mammals
18.
Brain Sci ; 13(3)2023 Feb 26.
Article in English | MEDLINE | ID: mdl-36979214

ABSTRACT

Optic nerve damage is a common cause of blindness. Optic nerve injury is often accompanied by fundus vascular disease, retinal ganglion cell apoptosis, and changes in retinal thickness. These changes can cause alterations in protein expression within neurons in the retina. Proteomics analysis offers conclusive evidence to decode a biological system. Furthermore, animal models of optic nerve injury made it possible to gain insight into pathological mechanisms, therapeutic targets, and effective treatment of such injuries. Proteomics takes the proteome as the research object and studies protein changes in cells and tissues. At present, a variety of proteomic analysis methods have been widely used in the research of optic nerve injury diseases. This review summarizes the application of proteomic research in optic nerve injury diseases and animal models of optic nerve injury. Additionally, differentially expressed proteins are summarized and analyzed. Various optic nerve injuries, including those associated with different etiologies, are discussed along with their potential therapeutic targets and future directions.

19.
J Nutr Biochem ; 117: 109333, 2023 07.
Article in English | MEDLINE | ID: mdl-36965783

ABSTRACT

Current clinical treatments have not yet effectively cured progressive retinal ganglion cell (RGC) death and axonal degeneration after optic nerve (ON) injury. We previously demonstrated green tea extract (GTE) can reduce RGC death in rats after ischemic injury. Here, we aim to determine the prophylactic and therapeutic effects and mechanisms of GTE on RGC survival and axonal regeneration in rats with ON injury. GTE (275 or 550 mg/kg) was administered intragastrically for 7 d before or 14 d post-ON crush surgery in adult Fischer 344 rats. Rats with pre- or post-operative treatment of 275 mg/kg GTE showed significantly higher numbers of RGCs and regenerated axons post-ON injury with improved pupillary light reflex as compared to saline-treated rats. Akt and Erk p42/44 activation was higher in the retina of rats given 275 mg/kg GTE pre-surgery, whereas Stat3 activation was higher in those with 275 mg/kg GTE post-operation. Less activated microglia were observed in rats with pre-treatment of 275 or 550 mg/kg GTE. RNA sequencing analysis identified the downregulation of inflammation, apoptosis, and microglia activation genes in the retina of rats with pre- or post-treatment with 275 mg/kg GTE as compared to the saline-treated rats. In summary, this study revealed the prophylactic and therapeutic treatment effects of GTE on RGC survival and axonal regeneration in rats with ON injury, indicating a potential alternative treatment for traumatic optic neuropathy.


Subject(s)
Optic Nerve Injuries , Retinal Ganglion Cells , Rats , Animals , Retinal Ganglion Cells/metabolism , Optic Nerve Injuries/drug therapy , Optic Nerve Injuries/metabolism , Nerve Regeneration/physiology , Rats, Inbred F344 , Tea , Cell Survival
20.
Development ; 150(8)2023 04 15.
Article in English | MEDLINE | ID: mdl-36971369

ABSTRACT

Failure of central nervous system projection neurons to spontaneously regenerate long-distance axons underlies irreversibility of white matter pathologies. A barrier to axonal regenerative research is that the axons regenerating in response to experimental treatments stall growth before reaching post-synaptic targets. Here, we test the hypothesis that the interaction of regenerating axons with live oligodendrocytes, which were absent during developmental axon growth, contributes to stalling axonal growth. To test this hypothesis, first, we used single cell RNA-seq (scRNA-seq) and immunohistology to investigate whether post-injury born oligodendrocytes incorporate into the glial scar after optic nerve injury. Then, we administered demyelination-inducing cuprizone and stimulated axon regeneration by Pten knockdown (KD) after optic nerve crush. We found that post-injury born oligodendrocyte lineage cells incorporate into the glial scar, where they are susceptible to the demyelination diet, which reduced their presence in the glial scar. We further found that the demyelination diet enhanced Pten KD-stimulated axon regeneration and that localized cuprizone injection promoted axon regeneration. We also present a resource for comparing the gene expression of scRNA-seq-profiled normal and injured optic nerve oligodendrocyte lineage cells.


Subject(s)
Axons , Demyelinating Diseases , Humans , Axons/physiology , Gliosis/metabolism , Gliosis/pathology , Cuprizone , Nerve Regeneration/physiology , Retinal Ganglion Cells/metabolism , Oligodendroglia , Demyelinating Diseases/chemically induced , Demyelinating Diseases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...