1
|
Goldberg JL. Bringing Eye Transplant Into the Light. JAMA 2024; 332:1523-1524. [PMID: 39250112 DOI: 10.1001/jama.2024.14811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/10/2024]
Affiliation(s)
- Jeffrey L Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, California
| |
Collapse
|
2
|
Wood EH, Kreymerman A, Kowal T, Buickians D, Sun Y, Muscat S, Mercola M, Moshfeghi DM, Goldberg JL. Cellular and subcellular optogenetic approaches towards neuroprotection and vision restoration. Prog Retin Eye Res 2023; 96:101153. [PMID: 36503723 PMCID: PMC10247900 DOI: 10.1016/j.preteyeres.2022.101153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 11/27/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022]
Abstract
Optogenetics is defined as the combination of genetic and optical methods to induce or inhibit well-defined events in isolated cells, tissues, or animals. While optogenetics within ophthalmology has been primarily applied towards treating inherited retinal disease, there are a myriad of other applications that hold great promise for a variety of eye diseases including cellular regeneration, modulation of mitochondria and metabolism, regulation of intraocular pressure, and pain control. Supported by primary data from the authors' work with in vitro and in vivo applications, we introduce a novel approach to metabolic regulation, Opsins to Restore Cellular ATP (ORCA). We review the fundamental constructs for ophthalmic optogenetics, present current therapeutic approaches and clinical trials, and discuss the future of subcellular and signaling pathway applications for neuroprotection and vision restoration.
Collapse
Affiliation(s)
- Edward H Wood
- Spencer Center for Vision Research, Byers Eye Institute, Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA; Stanford Cardiovascular Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Alexander Kreymerman
- Spencer Center for Vision Research, Byers Eye Institute, Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA; Stanford Cardiovascular Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Tia Kowal
- Spencer Center for Vision Research, Byers Eye Institute, Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - David Buickians
- Spencer Center for Vision Research, Byers Eye Institute, Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Yang Sun
- Spencer Center for Vision Research, Byers Eye Institute, Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Stephanie Muscat
- Spencer Center for Vision Research, Byers Eye Institute, Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Mark Mercola
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Darius M Moshfeghi
- Spencer Center for Vision Research, Byers Eye Institute, Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Jeffrey L Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA.
| |
Collapse
|
3
|
Pacwa A, Machowicz J, Akhtar S, Rodak P, Liu X, Pietrucha-Dutczak M, Lewin-Kowalik J, Amadio M, Smedowski A. Deficiency of the RNA-binding protein ELAVL1/HuR leads to the failure of endogenous and exogenous neuroprotection of retinal ganglion cells. Front Cell Neurosci 2023; 17:1131356. [PMID: 36874215 PMCID: PMC9982123 DOI: 10.3389/fncel.2023.1131356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 02/06/2023] [Indexed: 02/19/2023] Open
Abstract
Introduction ELAVL1/HuR is a keystone regulator of gene expression at the posttranscriptional level, including stress response and homeostasis maintenance. The aim of this study was to evaluate the impact of hur silencing on the age-related degeneration of retinal ganglion cells (RGC), which potentially describes the efficiency of endogenous neuroprotection mechanisms, as well as to assess the exogenous neuroprotection capacity of hur-silenced RGC in the rat glaucoma model. Methods The study consisted of in vitro and in vivo approaches. In vitro, we used rat B-35 cells to investigate, whether AAV-shRNA-HuR delivery affects survival and oxidative stress markers under temperature and excitotoxic insults. In vivo approach consisted of two different settings. In first one, 35 eight-week-old rats received intravitreal injection of AAV-shRNA-HuR or AAV-shRNA scramble control. Animals underwent electroretinography tests and were sacrificed 2, 4 or 6 months after injection. Retinas and optic nerves were collected and processed for immunostainings, electron microscopy and stereology. For the second approach, animals received similar gene constructs. To induce chronic glaucoma, 8 weeks after AAV injection, unilateral episcleral vein cauterization was performed. Animals from each group received intravitreal injection of metallothionein II. Animals underwent electroretinography tests and were sacrificed 8 weeks later. Retinas and optic nerves were collected and processed for immunostainings, electron microscopy and stereology. Results Silencing of hur induced apoptosis and increased oxidative stress markers in B-35 cells. Additionally, shRNA treatment impaired the cellular stress response to temperature and excitotoxic insults. In vivo, RGC count was decreased by 39% in shRNA-HuR group 6 months after injection, when compared to shRNA scramble control group. In neuroprotection study, the average loss of RGCs was 35% in animals with glaucoma treated with metallothionein and shRNA-HuR and 11.4% in animals with glaucoma treated with metallothionein and the scramble control shRNA. An alteration in HuR cellular content resulted in diminished photopic negative responses in the electroretinogram. Conclusions Based on our findings, we conclude that HuR is essential for the survival and efficient neuroprotection of RGC and that the induced alteration in HuR content accelerates both the age-related and glaucoma-induced decline in RGC number and function, further confirming HuR's key role in maintaining cell homeostasis and its possible involvement in the pathogenesis of glaucoma.
Collapse
Affiliation(s)
- Anna Pacwa
- Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
- GlaucoTech Co., Katowice, Poland
| | - Joanna Machowicz
- Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Saeed Akhtar
- College of Applied Medical Sciences, Inaya Medical Colleges, Riyadh, Saudi Arabia
- Department of Optometry, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Piotr Rodak
- Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Xiaonan Liu
- Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Marita Pietrucha-Dutczak
- Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Joanna Lewin-Kowalik
- Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
- GlaucoTech Co., Katowice, Poland
| | - Marialaura Amadio
- Department of Drug Sciences, Section of Pharmacology, The University of Pavia, Pavia, Italy
| | - Adrian Smedowski
- Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
- GlaucoTech Co., Katowice, Poland
| |
Collapse
|
4
|
Wang X, Pang J, Cui J, Liu A, Wang H. Inhibition of microRNA-19a-3p alleviates the neuropathic pain (NP) in rats after chronic constriction injury (CCI) via targeting KLF7. Transpl Immunol 2023; 76:101735. [PMID: 36334791 DOI: 10.1016/j.trim.2022.101735] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 09/19/2022] [Accepted: 10/30/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND/PURPOSE Neuropathic pain(NP) is derived from the dysfunctions of nerve system. The current research is to explore the impact and mechanism of miR-19a-3p in neuropathic pain in rats. METHODS The NP was induced through the chronic constriction injury (CCI) surgery in rats. The pro-inflammatory factors (IL-1β, IL-6, TNF-α) in spinal cord tissues from rats were measured using Elisa kits. Moreover, the different levels of thermal hyperalgesia and mechanical allodynia in rats were examined through paw withdrawal latency (PWL) and paw withdrawal threshold (PWT). To investigate into the role of miR-19a-3p and KLF7 in NP of rats, the knockdown of miR-19a-3p alone or along with KLF7 downregulation in rats were achieved through lentivirus injection. The miR-19a-3p and KLF7 expression in spinal cord of rats on Day 3,7,14 after CCI were detected using RT-qPCR. The protein expression of KLF7 were measured by Western blot. Bioinformatics and luciferase assays were used for the prediction and verification of bindings between KLF7 and miR-19a-3p. RESULTS CCI surgery caused neuropathic pain in rats with the levels of inflammatory cytokines increased and PWL and PWT decreased. Moreover, miR-19a-3p expression was increased while the protein and mRNA levels were decreased in spinal cord tissues in rats after CCI surgery. In rat microglial cells, miR-19a-3p downregulation could promote the KLF7 in both mRNA and protein expression. In spinal cord tissues of rats, the inhibition of miR-19a-3p enhanced the KLF7 expression. Furthermore, miR-19a-3p downregulation suppressed the IL-1β, IL-6 and TNF-α concentrations, and could decrease the NP but inhibition of KLF7 could partially reverse this in CCI rats. CONCLUSION miR-19a-3p inhibition may alleviate NP via KLF7 in CCI rats.
Collapse
Affiliation(s)
- Xiaopeng Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China.
| | - Jun Pang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China
| | - Jian Cui
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin 300100, China
| | - Aifen Liu
- Department of Anesthesiology, the Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Hui Wang
- Department of General Surgery, Tianjin Public Security Hospital, Tianjin 300042, China
| |
Collapse
|
5
|
Song Y, Wang M, Zhao S, Tian Y, Zhang C. Matrine promotes mitochondrial biosynthesis and reduces oxidative stress in experimental optic neuritis. Front Pharmacol 2022; 13:936632. [PMID: 36238552 PMCID: PMC9552203 DOI: 10.3389/fphar.2022.936632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Optic neuritis (ON), characterized by inflammation of the optic nerve and apoptosis of retinal ganglion cells (RGCs), is one of the leading causes of blindness in patients. Given that RGC, as an energy-intensive cell, is vulnerable to mitochondrial dysfunction, improving mitochondrial function and reducing oxidative stress could protect these cells. Matrine (MAT), an alkaloid derived from Sophoraflavescens, has been shown to regulate immunity and protect neurons in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis and ON. However, the protective mechanism of MAT on RGCs is largely unknown. In this study, we show that MAT treatment significantly reduced the degree of inflammatory infiltration and demyelination of the optic nerve and increased the survival rate of RGCs. The expression of Sirtuin 1 (SIRT1), a member of an evolutionarily conserved gene family (sirtuins), was upregulated, as well as its downstream molecules Nrf2 and PGC-1α. The percentage of TOMM20-positive cells was also increased remarkably in RGCs after MAT treatment. Thus, our results indicate that MAT protects RGCs from apoptosis, at least in part, by activating SIRT1 to regulate PGC-1α and Nrf2, which, together, promote mitochondrial biosynthesis and reduce the oxidative stress of RGCs.
Collapse
Affiliation(s)
- Yifan Song
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Mengru Wang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Suyan Zhao
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Yanjie Tian
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
- *Correspondence: Yanjie Tian,
| | - Chun Zhang
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| |
Collapse
|
6
|
Zhang Q, Li Y, Zhuo Y. Synaptic or Non-synaptic? Different Intercellular Interactions with Retinal Ganglion Cells in Optic Nerve Regeneration. Mol Neurobiol 2022; 59:3052-3072. [PMID: 35266115 PMCID: PMC9016027 DOI: 10.1007/s12035-022-02781-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 02/24/2022] [Indexed: 12/31/2022]
Abstract
Axons of adult neurons in the mammalian central nervous system generally fail to regenerate by themselves, and few if any therapeutic options exist to reverse this situation. Due to a weak intrinsic potential for axon growth and the presence of strong extrinsic inhibitors, retinal ganglion cells (RGCs) cannot regenerate their axons spontaneously after optic nerve injury and eventually undergo apoptosis, resulting in permanent visual dysfunction. Regarding the extracellular environment, research to date has generally focused on glial cells and inflammatory cells, while few studies have discussed the potentially significant role of interneurons that make direct connections with RGCs as part of the complex retinal circuitry. In this study, we provide a novel angle to summarize these extracellular influences following optic nerve injury as "intercellular interactions" with RGCs and classify these interactions as synaptic and non-synaptic. By discussing current knowledge of non-synaptic (glial cells and inflammatory cells) and synaptic (mostly amacrine cells and bipolar cells) interactions, we hope to accentuate the previously neglected but significant effects of pre-synaptic interneurons and bring unique insights into future pursuit of optic nerve regeneration and visual function recovery.
Collapse
Affiliation(s)
- Qi Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Yiqing Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China.
| | - Yehong Zhuo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China.
| |
Collapse
|
7
|
Retinal ganglion cell loss in an ex vivo mouse model of optic nerve cut is prevented by curcumin treatment. Cell Death Discov 2021; 7:394. [PMID: 34911931 PMCID: PMC8674341 DOI: 10.1038/s41420-021-00760-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/27/2021] [Accepted: 11/09/2021] [Indexed: 11/08/2022] Open
Abstract
Retinal ganglion cell (RGC) loss is a pathologic feature common to several retinopathies associated to optic nerve damage, leading to visual loss and blindness. Although several scientific efforts have been spent to understand the molecular and cellular changes occurring in retinal degeneration, an effective therapy to counteract the retinal damage is still not available. Here we show that eyeballs, enucleated with the concomitant optic nerve cut (ONC), when kept in PBS for 24 h showed retinal and optic nerve degeneration. Examining retinas and optic nerves at different time points in a temporal window of 24 h, we found a thinning of some retinal layers especially RGC's layer, observing a powerful RGC loss after 24 h correlated with an apoptotic, MAPKs and degradative pathways dysfunctions. Specifically, we detected a time-dependent increase of Caspase-3, -9 and pro-apoptotic marker levels, associated with a strong reduction of BRN3A and NeuN levels. Importantly, a powerful activation of JNK, c-Jun, and ERK signaling (MAPKs) were observed, correlated with a significant augmented SUMO-1 and UBC9 protein levels. The degradation signaling pathways was also altered, causing a significant decrease of ubiquitination level and an increased LC3B activation. Notably, it was also detected an augmented Tau protein level. Curcumin, a powerful antioxidant natural compound, prevented the alterations of apoptotic cascade, MAPKs, and SUMO-1 pathways and the degradation system, preserving the RGC survival and the retinal layer thickness. This ex vivo retinal degeneration model could be a useful method to study, in a short time window, the effect of neuroprotective tools like curcumin that could represent a potential treatment to contrast retinal cell death.
Collapse
|
8
|
Coco-Martin RM, Pastor-Idoate S, Pastor JC. Cell Replacement Therapy for Retinal and Optic Nerve Diseases: Cell Sources, Clinical Trials and Challenges. Pharmaceutics 2021; 13:pharmaceutics13060865. [PMID: 34208272 PMCID: PMC8230855 DOI: 10.3390/pharmaceutics13060865] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 06/05/2021] [Accepted: 06/07/2021] [Indexed: 12/15/2022] Open
Abstract
The aim of this review was to provide an update on the potential of cell therapies to restore or replace damaged and/or lost cells in retinal degenerative and optic nerve diseases, describing the available cell sources and the challenges involved in such treatments when these techniques are applied in real clinical practice. Sources include human fetal retinal stem cells, allogenic cadaveric human cells, adult hippocampal neural stem cells, human CNS stem cells, ciliary pigmented epithelial cells, limbal stem cells, retinal progenitor cells (RPCs), human pluripotent stem cells (PSCs) (including both human embryonic stem cells (ESCs) and human induced pluripotent stem cells (iPSCs)) and mesenchymal stem cells (MSCs). Of these, RPCs, PSCs and MSCs have already entered early-stage clinical trials since they can all differentiate into RPE, photoreceptors or ganglion cells, and have demonstrated safety, while showing some indicators of efficacy. Stem/progenitor cell therapies for retinal diseases still have some drawbacks, such as the inhibition of proliferation and/or differentiation in vitro (with the exception of RPE) and the limited long-term survival and functioning of grafts in vivo. Some other issues remain to be solved concerning the clinical translation of cell-based therapy, including (1) the ability to enrich for specific retinal subtypes; (2) cell survival; (3) cell delivery, which may need to incorporate a scaffold to induce correct cell polarization, which increases the size of the retinotomy in surgery and, therefore, the chance of severe complications; (4) the need to induce a localized retinal detachment to perform the subretinal placement of the transplanted cell; (5) the evaluation of the risk of tumor formation caused by the undifferentiated stem cells and prolific progenitor cells. Despite these challenges, stem/progenitor cells represent the most promising strategy for retinal and optic nerve disease treatment in the near future, and therapeutics assisted by gene techniques, neuroprotective compounds and artificial devices can be applied to fulfil clinical needs.
Collapse
Affiliation(s)
- Rosa M. Coco-Martin
- Instituto de Oftalmobiologia Aplicada (IOBA), Medical School, Universidad de Valladolid, 47011 Valladolid, Spain; (S.P.-I.); (J.C.P.)
- National Institute of Health Carlos III (ISCIII), (RETICS) Cooperative Health Network for Research in Ophthalmology (Oftared), 28040 Madrid, Spain
- Correspondence: ; Tel.: +34-983423559
| | - Salvador Pastor-Idoate
- Instituto de Oftalmobiologia Aplicada (IOBA), Medical School, Universidad de Valladolid, 47011 Valladolid, Spain; (S.P.-I.); (J.C.P.)
- National Institute of Health Carlos III (ISCIII), (RETICS) Cooperative Health Network for Research in Ophthalmology (Oftared), 28040 Madrid, Spain
- Department of Ophthalmology, Hospital Clinico Universitario of Valladolid, 47003 Valladolid, Spain
| | - Jose Carlos Pastor
- Instituto de Oftalmobiologia Aplicada (IOBA), Medical School, Universidad de Valladolid, 47011 Valladolid, Spain; (S.P.-I.); (J.C.P.)
- National Institute of Health Carlos III (ISCIII), (RETICS) Cooperative Health Network for Research in Ophthalmology (Oftared), 28040 Madrid, Spain
- Department of Ophthalmology, Hospital Clinico Universitario of Valladolid, 47003 Valladolid, Spain
- Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Fundacion del Instituto de Estudios de Ciencias de la Salud de Castilla y León (ICSCYL), 42002 Soria, Spain
| |
Collapse
|
9
|
Shao W, Liu X, Gao L, Tian C, Shi Q. αA-Crystallin inhibits optic nerve astrocyte activation induced by oxygen-glucose deprivation in vitro. Life Sci 2021; 278:119533. [PMID: 33887346 DOI: 10.1016/j.lfs.2021.119533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 04/07/2021] [Accepted: 04/15/2021] [Indexed: 11/25/2022]
Abstract
AIMS A previous study reported that intravitreal injection of αA-crystallin inhibits glial scar formation after optic nerve traumatic injury. The purpose of this study was to investigate the effect of αA-crystallin on optic nerve astrocytes induced by oxygen glucose deprivation (OGD) in vitro. MATERIALS AND METHODS Optic nerve astrocytes from newborn Long Evans rats were cultured with αA-crystallin (10-4 g/l) to detect the effects of αA-crystallin on astrocytes. Using a scratch assay, the effect of αA-crystallin treatment on astrocyte migration was assessed. Astrocytes were exposed to OGD and glucose reintroduction/reoxygenation culture for 24 h and 48 h. The expression of glial fibrillary acidic protein (GFAP) and neurocan were subsequently evaluated via immunocytochemistry and western blot. BMP2/4, BMPRIa/Ib and Smad1/5/8 mRNA expression levels were detected by RT-PCR. KEY FINDINGS The results showed that αA-crystallin slowed the migration of astrocytes in filling the scratch gaps. GFAP and neurocan expression in astrocytes was increased after OGD. However, after treatment with αA-crystallin, GFAP and neurocan expression levels clearly decreased. Furthermore, RT-PCR showed that BMP2 and BMP4 mRNA expression levels decreased significantly. SIGNIFICANCE These results suggest that αA-crystallin inhibits the activation of astrocytes after OGD injury in vitro. Inhibition of the BMP/Smad signaling pathway might be the mechanism underlying this effect.
Collapse
Affiliation(s)
- Weiyang Shao
- Ophthalmology Department, Sixth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Xiao Liu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing 400038, China
| | - Lixiong Gao
- Ophthalmology Department, Sixth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Chunyu Tian
- Ophthalmology Department, Sixth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Qian Shi
- Ophthalmology Department, Sixth Medical Center of PLA General Hospital, Beijing 100048, China.
| |
Collapse
|
10
|
Rezaei S, Dabirmanesh B, Zare L, Golestani A, Javan M, Khajeh K. Enhancing myelin repair in experimental model of multiple sclerosis using immobilized chondroitinase ABC I on porous silicon nanoparticles. Int J Biol Macromol 2019; 146:162-170. [PMID: 31899243 DOI: 10.1016/j.ijbiomac.2019.12.258] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/19/2019] [Accepted: 12/30/2019] [Indexed: 12/27/2022]
Abstract
Removal of chondroitin sulfate proteoglycans (CSPGs) with chondroitinase ABC I (ChABC) facilitates axonal plasticity, axonal regeneration and remyelination, following injury to the central nervous system (CNS). However, the ChABC rapidly undergoes thermal inactivity and needs to be injected repeatedly. Here this limitation was overcame by immobilizing the ChABC on porous silicon (PSi) nanoparticles (ChABC@PSi). The efficacy of immobilized ChABC on CSPGs level and the demyelination insult was assessed in mice corpora callosa demyelinated by 6 weeks cuprizone (CPZ) feeding. ChABC@PSi was able to reduce the amount of CSPGs two weeks after animals treatment. CSPGs digestion by ChABC@PSi reduced the extent of demyelinated area as well as the astrogliosis. Furthermore, ChABC@PSi treatment increased the number of newly generated oligodendrocyte lineage cells which imply for enhanced myelin repair. Our results showed that effective CSPGs digestion by ChABC@PSi enhanced remyelination in CPZ model. Accordingly, ChABC@PSi may have a great potential to be used for treatment of diseases like multiple sclerosis and spinal cord injury by promoting the regeneration of damaged nerves.
Collapse
Affiliation(s)
- Safoura Rezaei
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Bahareh Dabirmanesh
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Leila Zare
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Abolfazl Golestani
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Khosro Khajeh
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran; Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
11
|
Zhang J, Liu W, Zhang X, Lin S, Yan J, Ye J. Sema3A inhibits axonal regeneration of retinal ganglion cells via ROCK2. Brain Res 2019; 1727:146555. [PMID: 31733191 DOI: 10.1016/j.brainres.2019.146555] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 11/03/2019] [Accepted: 11/12/2019] [Indexed: 01/04/2023]
Abstract
Successful regeneration of injured axons in the adult mammalian central nervous system (CNS) is mainly limited by lesion-induced neuronal apoptosis and the inhibitory environment consisting of numerous extrinsic and intrinsic factors. Semaphorin 3A (Sema3A), a classic axonal guidance cue, contributes to the failure of axonal regeneration and can be neutralized to enhance axonal regeneration. Previous studies have suggested that blockage of rho-associated protein kinase 2 (ROCK2) also exerts a protective effect on the survival and axonal regeneration of retinal ganglion cells (RGC, RGCs) after injury. Yet unresolved question is the interaction between the two factors. We thus evaluated the role of Sema3A and ROCK2 in RGC axonal regeneration. In this study, we first examined the expression of Sema3A and ROCK2 against optic nerve crush in vivo and oxygen-glucose deprivation insult to RGCs in vitro at different time points. Then Sema3A, ROCK2 inhibitor Y-27632, combination of both and phosphate-buffered saline (PBS) only were injected into the vitreous cavity after optic nerve crush at various times in different experiments. In order to assess axonal regeneration, we detected the mRNA levels of small proline-rich protein 1A (Sprr1A) and growth-associated protein 43 (GAP43) by quantitative real time-polymerase chain reaction (RT-qPCR), evaluated visual function by Flash Visual Evoked Potentials (F-VEPs), and checked the protein level of GAP43 by immunofluorescent staining. Our results demonstrated that Sema3A significantly suppressed optic nerve regeneration and this effect can be attenuated via blocking ROCK2. Moreover, Sema3A promoted the phosphorylation of myosin light chain 2 (MLC2) (specific downstream effector of ROCK2 concerning neurite growth). Collectively, Sema3A may negatively regulate axonal regeneration through ROCK2 in RGCs.
Collapse
Affiliation(s)
- Jieqiong Zhang
- Department of Ophthalmology, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Wenyi Liu
- Department of Ophthalmology, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Xi Zhang
- Department of Ophthalmology, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Sen Lin
- Department of Ophthalmology, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Jun Yan
- Department 1, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China.
| | - Jian Ye
- Department of Ophthalmology, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China.
| |
Collapse
|
12
|
Immunological considerations and concerns as pertinent to whole eye transplantation. Curr Opin Organ Transplant 2019; 24:726-732. [PMID: 31689262 DOI: 10.1097/mot.0000000000000713] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
PURPOSE OF REVIEW The advent of clinical vascularized composite allotransplantation (VCA), offers hope for whole eye transplantation (WET) in patients with devastating vison loss that fails or defies current treatment options. Optic nerve regeneration and reintegration remain the overarching hurdles to WET. However, the realization of WET may indeed be limited by our lack of understanding of the singular immunological features of the eye as pertinent to graft survival and functional vision restoration in the setting of transplantation. RECENT FINDINGS Like other VCA, such as the hand or face, the eye includes multiple tissues with distinct embryonic lineage and differential antigenicity. The ultimate goal of vision restoration through WET requires optimal immune modulation of the graft for successful optic nerve regeneration. Our team is exploring barriers to our understanding of the immunology of the eye in the context of WET including the role of immune privilege and lymphatic drainage on rejection, as well as the effects ischemia, reperfusion injury and rejection on optic nerve regeneration. SUMMARY Elucidation of the unique immunological responses in the eye and adnexa after WET will provide foundational clues that will help inform therapies that prevent immune rejection without hindering optic nerve regeneration or reintegration.
Collapse
|
13
|
Demonstration of technical feasibility and viability of whole eye transplantation in a rodent model. J Plast Reconstr Aesthet Surg 2019; 72:1640-1650. [PMID: 31377202 DOI: 10.1016/j.bjps.2019.05.042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 04/17/2019] [Accepted: 05/02/2019] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Whole eye transplantation (WET) holds promise for vision restoration in devastating/disabling visual loss (congenital or traumatic) not amenable to surgical or neuroprosthetic treatment options. The eye includes multiple tissues with distinct embryonic lineage and differential antigenicity. Anatomically and immunologically, the eye is unique due to its avascular (cornea) and highly vascular (retina) components. Our goal was to establish technical feasibility, demonstrate graft viability, and evaluate histologic changes in ocular tissues/adnexae in a novel experimental model of WET that included globe, adnexal, optic nerve (ON), and periorbital soft tissues. METHODS Outbred Sprague-Dawley rats (n = 5) received heterotopic vascularized WET from donors. Each WET included the entire globe, adnexa, ON, and periorbital soft tissues supplied by the common carotid artery and external jugular vein. Viability and perfusion were confirmed by clinical examination, angiography and magnetic resonance imaging (MRI). Globe, adnexal, and periorbital tissues were analyzed for histopathologic changes, and the ON was examined for neuro-regeneration at study endpoint (30 days) or Banff Grade 3 rejection in the periorbital skin (whichever was earlier). RESULTS Gross examination confirmed transplant viability and corneal transparency. Average operative duration was 64.0 ± 5.8 min. Average ischemia time was 26.0 ± 4.2 min. MRI revealed loss of globe volume by 36.0 ± 2.8% after transplantation. Histopathology of globe and adnexal tissues showed unique and differential patterns of inflammatory cell infiltration. The ON revealed a neurodegeneration pattern. CONCLUSION The present study is the first in the literature to establish an experimental model of WET. This model holds significant potential in investigating mechanistic pathways, monitoring strategies or developing management approaches involving ocular viability, immune rejection, and ON regeneration after WET.
Collapse
|
14
|
Herrera E, Agudo-Barriuso M, Murcia-Belmonte V. Cranial Pair II: The Optic Nerves. Anat Rec (Hoboken) 2018; 302:428-445. [DOI: 10.1002/ar.23922] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 04/19/2017] [Accepted: 05/14/2017] [Indexed: 11/12/2022]
Affiliation(s)
- Eloísa Herrera
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH); Av. Santiago Ramón y Cajal, s/n., 03550 Sant Joan d'Alacant Alicante Spain
| | - Marta Agudo-Barriuso
- Departamento de Oftalmología, Facultad de Medicina; Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca (IMIB-Arrixaca); Murcia Spain
| | - Verónica Murcia-Belmonte
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH); Av. Santiago Ramón y Cajal, s/n., 03550 Sant Joan d'Alacant Alicante Spain
| |
Collapse
|
15
|
Stern JH, Tian Y, Funderburgh J, Pellegrini G, Zhang K, Goldberg JL, Ali RR, Young M, Xie Y, Temple S. Regenerating Eye Tissues to Preserve and Restore Vision. Cell Stem Cell 2018; 22:834-849. [PMID: 29859174 PMCID: PMC6492284 DOI: 10.1016/j.stem.2018.05.013] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ocular regenerative therapies are on track to revolutionize treatment of numerous blinding disorders, including corneal disease, cataract, glaucoma, retinitis pigmentosa, and age-related macular degeneration. A variety of transplantable products, delivered as cell suspensions or as preformed 3D structures combining cells and natural or artificial substrates, are in the pipeline. Here we review the status of clinical and preclinical studies for stem cell-based repair, covering key eye tissues from front to back, from cornea to retina, and including bioengineering approaches that advance cell product manufacturing. While recognizing the challenges, we look forward to a deep portfolio of sight-restoring, stem cell-based medicine. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Jeffrey H Stern
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA; Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA
| | - Yangzi Tian
- Colleges of Nanoscale Science and Engineering, SUNY Polytechnic Institute, 257 Fuller Road, Albany, NY 12203, USA
| | - James Funderburgh
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Graziella Pellegrini
- Centre for Regenerative Medicine, University of Modena and Reggio Emilia, via G.Gottardi 100, 41125 Modena, Italy
| | - Kang Zhang
- Shiley Eye Institute and Institute for Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University and Guangzhou Regenerative Medicine and Health Laboratory, Guangzhou 510060, China
| | - Jeffrey L Goldberg
- Byers Eye Institute at Stanford University, 2452 Watson Court, Palo Alto, CA 94303, USA
| | - Robin R Ali
- Department of Genetics, University College London Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK; NIHR Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, City Road, London EC1V 2PD, UK; Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA
| | - Michael Young
- The Schepens Eye Research Institute, Massachusetts Eye and Ear, an affiliate of Harvard Medical School, Boston, MA 02114, USA
| | - Yubing Xie
- Colleges of Nanoscale Science and Engineering, SUNY Polytechnic Institute, 257 Fuller Road, Albany, NY 12203, USA
| | - Sally Temple
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA; Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA.
| |
Collapse
|
16
|
Wang F, Dang Y, Wang J, Zhou T, Zhu Y. Gypenosides attenuate lipopolysaccharide-induced optic neuritis in rats. Acta Histochem 2018; 120:340-346. [PMID: 29559175 DOI: 10.1016/j.acthis.2018.03.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 03/05/2018] [Accepted: 03/06/2018] [Indexed: 12/12/2022]
Abstract
PURPOSE To evaluate the effect of gypenosides (GPs) on lipopolysaccharide (LPS)-induced optic neuritis rats. METHODS Optic neuritis was induced by a single microinjection of LPS into the optic nerve of Sprague Dawley rats. GPs (400 mg/kg) was administrated by gavage for 21 days. The optic nerve structure changes and demyelination were observed after hematoxylin & eosin and Luxol-fast blue staining. Apoptosis of retinal ganglion cells (RGCs) was evaluated using Brn3a-TUNEL double staining. Expression of CD68 and glial fibrillary acidic protein (GFAP) were detected using immunofluorescence staining. The mRNA levels of inflammatory factors were measured using quantitative real-time PCR. The protein expression levels in the signal transducer and activator of transcription (STAT) and nuclear factor-κB (NF-κB) pathways were detected using Western blot. RESULTS GPs treatment prevented the optic nerve structure changes and demyelination in the rats with optic neuritis. GPs treatment downregulated LPS-induced overexpressions of CD68, GFAP and pro-inflammatory factors. GPs treatment inhibited STAT1 and 3 phosphorylation and NF-κB nuclear translocation in the optic nerve and retina of rats with optic neuritis. CONCLUSION GPs attenuate LPS-induced inflammation, demyelination and optic nerve damage which may be associated with the inhibition of the NF-κB and STAT pathways.
Collapse
|
17
|
|
18
|
Inhibition of miR-21 ameliorates excessive astrocyte activation and promotes axon regeneration following optic nerve crush. Neuropharmacology 2018; 137:33-49. [PMID: 29709341 DOI: 10.1016/j.neuropharm.2018.04.028] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/18/2018] [Accepted: 04/26/2018] [Indexed: 12/15/2022]
Abstract
Optic nerve injury is a leading cause of irreversible visual impairment worldwide and can even cause blindness. Excessive activation of astrocytes has negative effects on the repair and recovery of retinal ganglion cells following optic nerve injury. However, the molecular and cellular mechanisms underlying astrocyte activation after optic nerve injury remain largely unknown. In the present study, we explored the effects of microRNA-21 (miR-21) on axon regeneration and flash visual evoked potential (F-VEP) and the underlying mechanisms of these effects based on astrocyte activation in the rat model of optic nerve crush (ONC). To the best of our knowledge, this article is the first to report that inhibition of miR-21 enhances axonal regeneration and promotes functional recovery in F-VEP in the rat model of ONC. Furthermore, inhibition of miR-21 attenuates excessive astrocyte activation and glial scar formation, thereby promoting axonal regeneration by regulating the epidermal growth factor receptor (EGFR) pathway. In addition, we observed that the expression of tissue inhibitor of metalloproteinase-3, a target gene of miR-21, was inhibited during this process. Taken together, these findings demonstrate that inhibition of miR-21 regulates the EGFR pathway, ameliorating excessive astrocyte activation and glial scar progression and promoting axonal regeneration and alleviating impairment in F-VEP function in a model of ONC. This study's results suggest that miR-21 may represent a therapeutic target for optic nerve injury.
Collapse
|
19
|
Li HJ, Sun ZL, Yang XT, Zhu L, Feng DF. Exploring Optic Nerve Axon Regeneration. Curr Neuropharmacol 2018; 15:861-873. [PMID: 28029073 PMCID: PMC5652030 DOI: 10.2174/1570159x14666161227150250] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Revised: 12/14/2016] [Accepted: 12/22/2016] [Indexed: 12/13/2022] Open
Abstract
Background: Traumatic optic nerve injury is a leading cause of irreversible blindness across the world and causes progressive visual impairment attributed to the dysfunction and death of retinal ganglion cells (RGCs). To date, neither pharmacological nor surgical interventions are sufficient to halt or reverse the progress of visual loss. Axon regeneration is critical for functional recovery of vision following optic nerve injury. After optic nerve injury, RGC axons usually fail to regrow and die, leading to the death of the RGCs and subsequently inducing the functional loss of vision. However, the detailed molecular mechanisms underlying axon regeneration after optic nerve injury remain poorly understood. Methods: Research content related to the detailed molecular mechanisms underlying axon regeneration after optic nerve injury have been reviewed. Results: The present review provides an overview of regarding potential strategies for axonal regeneration of RGCs and optic nerve repair, focusing on the role of cytokines and their downstream signaling pathways involved in intrinsic growth program and the inhibitory environment together with axon guidance cues for correct axon guidance. A more complete understanding of the factors limiting axonal regeneration will provide a rational basis, which contributes to develop improved treatments for optic nerve regeneration. These findings are encouraging and open the possibility that clinically meaningful regeneration may become achievable in the future. Conclusion: Combination of treatments towards overcoming growth-inhibitory molecules and enhancing intrinsic growth capacity combined with correct guidance using axon guidance cues is crucial for developing promising therapies to promote axon regeneration and functional recovery after ON injury.
Collapse
Affiliation(s)
- Hong-Jiang Li
- Department of Neurosurgery, No.9 People's Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 201999, China
| | - Zhao-Liang Sun
- Department of Neurosurgery, No.9 People's Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 201999, China
| | - Xi-Tao Yang
- Department of Neurosurgery, No.9 People's Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 201999, China
| | - Liang Zhu
- Department of Neurosurgery, No.9 People's Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 201999, China
| | - Dong-Fu Feng
- Department of Neurosurgery, No.9 People's Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 201999, China
| |
Collapse
|
20
|
Huang ZR, Chen HY, Hu ZZ, Xie P, Liu QH. PTEN knockdown with the Y444F mutant AAV2 vector promotes axonal regeneration in the adult optic nerve. Neural Regen Res 2018; 13:135-144. [PMID: 29451218 PMCID: PMC5840979 DOI: 10.4103/1673-5374.224381] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The lack of axonal regeneration is the major cause of vision loss after optic nerve injury in adult mammals. Activating the PI3K/AKT/mTOR signaling pathway has been shown to enhance the intrinsic growth capacity of neurons and to facilitate axonal regeneration in the central nervous system after injury. The deletion of the mTOR negative regulator phosphatase and tensin homolog (PTEN) enhances regeneration of adult corticospinal neurons and ganglion cells. In the present study, we used a tyrosine-mutated (Y444F) AAV2 vector to efficiently express a short hairpin RNA (shRNA) for silencing PTEN expression in retinal ganglion cells. We evaluated cell survival and axonal regeneration in a rat model of optic nerve axotomy. The rats received an intravitreal injection of wildtype AAV2 or Y444F mutant AAV2 (both carrying shRNA to PTEN) 4 weeks before optic nerve axotomy. Compared with the wildtype AAV2 vector, the Y444F mutant AAV2 vector enhanced retinal ganglia cell survival and stimulated axonal regeneration to a greater extent 6 weeks after axotomy. Moreover, post-axotomy injection of the Y444F AAV2 vector expressing the shRNA to PTEN rescued ~19% of retinal ganglion cells and induced axons to regenerate near to the optic chiasm. Taken together, our results demonstrate that PTEN knockdown with the Y444F AAV2 vector promotes retinal ganglion cell survival and stimulates long-distance axonal regeneration after optic nerve axotomy. Therefore, the Y444F AAV2 vector might be a promising gene therapy tool for treating optic nerve injury.
Collapse
Affiliation(s)
- Zheng-Ru Huang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing; Department of Ophthalmology, the Second People's Hospital of Changshu, Changshu, Jiangsu Province, China
| | - Hai-Ying Chen
- Department of Ophthalmology, The Second People's Hospital of Changshu, Changshu, Jiangsu Province, China
| | - Zi-Zhong Hu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ping Xie
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Qing-Huai Liu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
21
|
|
22
|
The use of induced pluripotent stem cells for studying and treating optic neuropathies. Curr Opin Organ Transplant 2017; 21:484-9. [PMID: 27517502 DOI: 10.1097/mot.0000000000000348] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE OF REVIEW The present review aims to provide an update of applications of induced pluripotent stem cells (iPSCs) for disease modeling, cell/gene therapy, and drug screening for optic neuropathies. RECENT FINDINGS Degeneration of retinal ganglion cells (RGCs) is a characteristic of optic neuropathies. Human iPSCs can serve as a model to investigate disease pathology and potential repair mechanisms. In recent years, significant progress has been made in generating RGCs from iPSCs. Various groups have reported the potential of iPSCs for modeling optic neuropathies, such as glaucoma. The literature also highlights the potential to use iPSC-derived cells for high-throughput drug and toxicity screening. SUMMARY The present review summarizes current work in the field of iPSCs in optic neuropathies. Future studies to characterize iPSC-derived RGCs in a more in-depth manner will help expand the use of iPSCs to model and treat optic neuropathic diseases. Furthermore, iPSC modeling can be used in drug development by offering a new avenue to test novel therapeutic drugs for optic neuropathies.
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW Recent advances in experimental studies of optic nerve regeneration to better understand the pathophysiology of axon regrowth and provide insights into the future treatment of numerous optic neuropathies. RECENT FINDINGS The optic nerve is part of the central nervous system and cannot regenerate if injured. There are several steps that regenerating axons of retinal ganglion cells (RGCs) must take following optic nerve injury that include: maximizing the intrinsic growth capacity of RGCs, overcoming the extrinsic growth-inhibitory environment of the optic nerve, and optimizing the reinnervation of regenerated axons to their targets in the brain. Recently, some degree of experimental optic nerve regeneration has been achieved by factors associated with inducing intraocular inflammation, providing exogenous neurotrophic factors, reactivating intrinsic growth capacity of mature RGCs, or by modifying the extrinsic growth-inhibitory environment of the optic nerve. In some experiments, regenerating axons have been shown to reinnervate their central targets in the brain. SUMMARY Further approaches to the combination of aforementioned treatments will be necessary to develop future therapeutic strategy to promote ultimate regeneration of the optic nerve and functional vision recovery after optic nerve injury.
Collapse
|
24
|
Huang Z, Hu Z, Xie P, Liu Q. Tyrosine-mutated AAV2-mediated shRNA silencing of PTEN promotes axon regeneration of adult optic nerve. PLoS One 2017; 12:e0174096. [PMID: 28323869 PMCID: PMC5360277 DOI: 10.1371/journal.pone.0174096] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 03/04/2017] [Indexed: 11/28/2022] Open
Abstract
Activating PI3K/AKT/mTOR signaling pathway via deleting phosphatase and tensin homolog (PTEN) has been confirmed to enhance intrinsic growth capacity of neurons to facilitate the axons regeneration of central nervous system after injury. Considering conditional gene deletion is currently not available in clinical practice, we exploited capsid residue tyrosine 444 to phenylalanine mutated single-stranded adeno-associated virus serotype 2 (AAV2) as a vector delivering short hairpin RNA to silence PTEN to promote retinal ganglion cells (RGCs) survival and axons regeneration in adult rat optic nerve axotomy paradigm. We found that mutant AAV2 displayed higher infection efficiency to RGCs and Müller cells by intravitreal injection, mediated PTEN suppression, resulted in much more RGCs survival and more robust axons regeneration compared with wild type AAV2, due to the different extent of the mTOR complex-1 activation and glutamate aspartate transporter (GLAST) regulation. These results suggest that high efficiency AAV2-mediated PTEN knockdown represents a practicable therapeutic strategy for optic neuropathy.
Collapse
Affiliation(s)
- ZhengRu Huang
- Department of Ophthalmology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- Department of Ophthalmology, the Second People´s Hospital of Changshu, Changshu, Jiangsu Province, China
| | - ZiZhong Hu
- Department of Ophthalmology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ping Xie
- Department of Ophthalmology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - QingHuai Liu
- Department of Ophthalmology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- * E-mail:
| |
Collapse
|
25
|
Patel AK, Park KK, Hackam AS. Wnt signaling promotes axonal regeneration following optic nerve injury in the mouse. Neuroscience 2016; 343:372-383. [PMID: 28011153 DOI: 10.1016/j.neuroscience.2016.12.020] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 12/07/2016] [Accepted: 12/12/2016] [Indexed: 02/08/2023]
Abstract
Adult mammalian CNS axons generally do not regenerate, creating an obstacle to effective repair and recovery after neuronal injury. The canonical Wnt/β-catenin signaling pathway is an essential signal transduction cascade that regulates axon growth and neurite extension in the developing mammalian embryo. In this study, we investigated whether a Wnt/β-catenin signaling activator could be repurposed to induce regeneration in the adult CNS after axonal injury. We used a retinal ganglion cell (RGC) axon crush injury model in a transgenic Wnt reporter mouse, and intravitreal injections were used to deliver Wnt3a or saline to the RGC cell bodies within the retina. Our findings demonstrated that Wnt3a induced Wnt signaling in RGCs and resulted in significant axonal regrowth past the lesion site when measured at two and four weeks post-injury. Furthermore, Wnt3a-injected eyes showed increased survival of RGCs and significantly higher pattern electroretinography (PERG) amplitudes compared to the control. Additionally, Wnt3a-induced axonal regeneration and RGC survival were associated with elevated activation of the transcription factor Stat3, and reducing expression of Stat3 using a conditional Stat3 knock-out mouse line led to diminished Wnt3a-dependent axonal regeneration and RGC survival. Therefore, these findings reveal a novel role for retinal Wnt signaling in axonal regrowth and RGC survival following axonal injury, which may lead to the development of novel therapies for axonal regeneration.
Collapse
Affiliation(s)
- Amit K Patel
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Kevin K Park
- Miami Project to Cure Paralysis, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Abigail S Hackam
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
26
|
Discussion: Total Human Eye Allotransplantation: Developing Surgical Protocols for Donor and Recipient Procedures. Plast Reconstr Surg 2016; 138:1309-1310. [PMID: 27879600 DOI: 10.1097/prs.0000000000002822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
27
|
Total Human Eye Allotransplantation: Developing Surgical Protocols for Donor and Recipient Procedures. Plast Reconstr Surg 2016; 138:1297-1308. [PMID: 27879599 DOI: 10.1097/prs.0000000000002821] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND Vascularized composite allotransplantation of the eye is an appealing, novel method for reconstruction of the nonfunctioning eye. The authors' group has established the first orthotopic model for eye transplantation in the rat. With advancements in immunomodulation strategies together with new therapies in neuroregeneration, parallel development of human surgical protocols is vital for ensuring momentum toward eye transplantation in actual patients. METHODS Cadaveric donor tissue harvest (n = 8) was performed with orbital exenteration, combined open craniotomy, and endonasal approach to ligate the ophthalmic artery with a cuff of paraclival internal carotid artery, for transection of the optic nerve at the optic chiasm and transection of cranial nerves III to VI and the superior ophthalmic vein at the cavernous sinus. Candidate recipient vessels (superficial temporal/internal maxillary/facial artery and superficial temporal/facial vein) were exposed. Vein grafts were required for all anastomoses. Donor tissue was secured in recipient orbits followed by sequential venous and arterial anastomoses and nerve coaptation. Pedicle lengths and calibers were measured. All steps were timed, photographed, video recorded, and critically analyzed after each operative session. RESULTS The technical feasibility of cadaveric donor procurement and transplantation to cadaveric recipient was established. Mean measurements included optic nerve length (39 mm) and caliber (5 mm), donor artery length (33 mm) and caliber (3 mm), and superior ophthalmic vein length (15 mm) and caliber (0.5 mm). Recipient superficial temporal, internal maxillary artery, and facial artery calibers were 0.8, 2, and 2 mm, respectively; and superior temporal and facial vein calibers were 0.8 and 2.5 mm, respectively. CONCLUSION This surgical protocol serves as a benchmark for optimization of technique, large-animal model development, and ultimately potentiating the possibility of vision restoration transplantation surgery. CLINICAL QUESTION/LEVEL OF EVIDENCE Therapeutic, V.
Collapse
|
28
|
Shao WY, Liu X, Gu XL, Ying X, Wu N, Xu HW, Wang Y. Promotion of axon regeneration and inhibition of astrocyte activation by alpha A-crystallin on crushed optic nerve. Int J Ophthalmol 2016; 9:955-66. [PMID: 27500100 DOI: 10.18240/ijo.2016.07.04] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Accepted: 05/25/2016] [Indexed: 11/23/2022] Open
Abstract
AIM To explore the effects of αA-crystallin in astrocyte gliosis after optic nerve crush (ONC) and the mechanism of α-crystallin in neuroprotection and axon regeneration. METHODS ONC was established on the Sprague-Dawley rat model and αA-crystallin (10(-4) g/L, 4 µL) was intravitreously injected into the rat model. Flash-visual evoked potential (F-VEP) was examined 14d after ONC, and the glial fibrillary acidic protein (GFAP) levels in the retina and crush site were analyzed 1, 3, 5, 7 and 14d after ONC by immunohistochemistry (IHC) and Western blot respectively. The levels of beta Tubulin (TUJ1), growth-associated membrane phosphoprotein-43 (GAP-43), chondroitin sulfate proteoglycans (CSPGs) and neurocan were also determined by IHC 14d after ONC. RESULTS GFAP level in the retina and the optic nerve significantly increased 1d after ONC, and reached the peak level 7d post-ONC. Injection of αA-crystallin significantly decreased GFAP level in both the retina and the crush site 3d after ONC, and induced astrocytes architecture remodeling at the crush site. Quantification of retinal ganglion cell (RGC) axons indicated αA-crystallin markedly promoted axon regeneration in ONC rats and enhanced the regenerated axons penetrated into the glial scar. CSPGs and neurocan expression also decreased 14d after αA-crystallin injection. The amplitude (N1-P1) and latency (P1) of F-VEP were also restored. CONCLUSION Our results suggest α-crystallin promotes the axon regeneration of RGCs and suppresses the activation of astrocytes.
Collapse
Affiliation(s)
- Wei-Yang Shao
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Xiao Liu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Xian-Liang Gu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Xi Ying
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Nan Wu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Hai-Wei Xu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Yi Wang
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| |
Collapse
|
29
|
Wang Y, Cameron EG, Li J, Stiles TL, Kritzer MD, Lodhavia R, Hertz J, Nguyen T, Kapiloff MS, Goldberg JL. Muscle A-Kinase Anchoring Protein-α is an Injury-Specific Signaling Scaffold Required for Neurotrophic- and Cyclic Adenosine Monophosphate-Mediated Survival. EBioMedicine 2015; 2:1880-7. [PMID: 26844267 PMCID: PMC4703706 DOI: 10.1016/j.ebiom.2015.10.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 10/17/2015] [Accepted: 10/22/2015] [Indexed: 12/11/2022] Open
Abstract
Neurotrophic factor and cAMP-dependent signaling promote the survival and neurite outgrowth of retinal ganglion cells (RGCs) after injury. However, the mechanisms conferring neuroprotection and neuroregeneration downstream to these signals are unclear. We now reveal that the scaffold protein muscle A-kinase anchoring protein-α (mAKAPα) is required for the survival and axon growth of cultured primary RGCs. Although genetic deletion of mAKAPα early in prenatal RGC development did not affect RGC survival into adulthood, nor promoted the death of RGCs in the uninjured adult retina, loss of mAKAPα in the adult increased RGC death after optic nerve crush. Importantly, mAKAPα was required for the neuroprotective effects of brain-derived neurotrophic factor and cyclic adenosine-monophosphate (cAMP) after injury. These results identify mAKAPα as a scaffold for signaling in the stressed neuron that is required for RGC neuroprotection after optic nerve injury. mAKAPα is a stress-specific mediator of RGC survival. mAKAP deletion does not affect RGC survival in development or in the uninjured adult retina. mAKAP is downregulated after optic nerve injury, and its further deletion exacerbates RGC death. mAKAP deletion suppresses the neuroprotective effects of cAMP and BDNF after injury.
After injury or in degenerative diseases, neurons of the central nervous system (CNS) fail to regenerate and often die partly due to a lack of pro-survival, trophic signaling. Better understanding of such signaling is important for the development of therapies that enhance survival and regeneration of neurons after injury. Here we identify a critical regulator of such signaling, mAKAPα, a scaffold protein that coordinates pro-survival signaling to enhance survival and regeneration in CNS neurons after injury. The neuroprotective role of mAKAPα will likely lead to further future insights into the detailed nature of survival signaling in adult neurons.
Collapse
Affiliation(s)
- Yan Wang
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Department of Ophthalmology, University of California, San Diego, CA 92093, United States
| | - Evan G Cameron
- Department of Ophthalmology, University of California, San Diego, CA 92093, United States; Byers Eye Institute, Stanford University, Palo Alto, CA 94303, United States
| | - Jinliang Li
- Department of Pediatrics, Interdisciplinary Stem Cell Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, United States; Department of Medicine, Interdisciplinary Stem Cell Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, United States
| | - Travis L Stiles
- Department of Ophthalmology, University of California, San Diego, CA 92093, United States
| | - Michael D Kritzer
- Department of Pediatrics, Interdisciplinary Stem Cell Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, United States; Department of Medicine, Interdisciplinary Stem Cell Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, United States
| | - Rahul Lodhavia
- Department of Ophthalmology, University of California, San Diego, CA 92093, United States
| | - Jonathan Hertz
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Tu Nguyen
- Department of Ophthalmology, University of California, San Diego, CA 92093, United States
| | - Michael S Kapiloff
- Department of Pediatrics, Interdisciplinary Stem Cell Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, United States; Department of Medicine, Interdisciplinary Stem Cell Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, United States
| | - Jeffrey L Goldberg
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Department of Ophthalmology, University of California, San Diego, CA 92093, United States; Byers Eye Institute, Stanford University, Palo Alto, CA 94303, United States
| |
Collapse
|
30
|
Human Pluripotent Stem Cell-Derived Retinal Ganglion Cells: Applications for the Study and Treatment of Optic Neuropathies. CURRENT OPHTHALMOLOGY REPORTS 2015; 3:200-206. [PMID: 26618076 DOI: 10.1007/s40135-015-0081-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
31
|
Chen M, Tian S, Glasgow NG, Gibson G, Yang X, Shiber CE, Funderburgh J, Watkins S, Johnson JW, Schuman JS, Liu H. Lgr5⁺ amacrine cells possess regenerative potential in the retina of adult mice. Aging Cell 2015; 14:635-43. [PMID: 25990970 PMCID: PMC4531077 DOI: 10.1111/acel.12346] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2015] [Indexed: 01/16/2023] Open
Abstract
Current knowledge indicates that the adult mammalian retina lacks regenerative capacity. Here, we show that the adult stem cell marker, leucine-rich repeat-containing G-protein-coupled receptor 5 (Lgr5), is expressed in the retina of adult mice. Lgr5+ cells are generated at late stages of retinal development and exhibit properties of differentiated amacrine interneurons (amacrine cells). Nevertheless, Lgr5+ amacrine cells contribute to regeneration of new retinal cells in the adult stage. The generation of new retinal cells, including retinal neurons and Müller glia from Lgr5+ amacrine cells, begins in early adulthood and continues as the animal ages. Together, these findings suggest that the mammalian retina is not devoid of regeneration as previously thought. It is rather dynamic, and Lgr5+ amacrine cells function as an endogenous regenerative source. The identification of such cells in the mammalian retina may provide new insights into neuronal regeneration and point to therapeutic opportunities for age-related retinal degenerative diseases.
Collapse
Affiliation(s)
- Mengfei Chen
- Department of Microbiology and Molecular Genetics University of Pittsburgh School of Medicine Pittsburgh PA USA
| | - Shenghe Tian
- Department of Ophthalmology and Visual Science Research Center University of Pittsburgh School of Medicine Pittsburgh PA USA
- Louis J. Fox Center for Vision Restoration of UPMC and the University of Pittsburgh Pittsburgh PA USA
| | - Nathan G. Glasgow
- Department of Neuroscience and Center for Neuroscience University of Pittsburgh Pittsburgh PA USA
| | - Gregory Gibson
- Center for Biologic Imaging University of Pittsburgh School of Medicine Pittsburgh PA USA
| | - Xiaoling Yang
- Department of Ophthalmology and Visual Science Research Center University of Pittsburgh School of Medicine Pittsburgh PA USA
| | - Christen E. Shiber
- Department of Neuroscience and Center for Neuroscience University of Pittsburgh Pittsburgh PA USA
| | - James Funderburgh
- Department of Ophthalmology and Visual Science Research Center University of Pittsburgh School of Medicine Pittsburgh PA USA
- Louis J. Fox Center for Vision Restoration of UPMC and the University of Pittsburgh Pittsburgh PA USA
- UPMC Eye Center Eye and Ear Institute Pittsburgh PA USA
| | - Simon Watkins
- Center for Biologic Imaging University of Pittsburgh School of Medicine Pittsburgh PA USA
- Department of Cell Biology and Physiology University of Pittsburgh School of Medicine Pittsburgh PA USA
| | - Jon W. Johnson
- Department of Neuroscience and Center for Neuroscience University of Pittsburgh Pittsburgh PA USA
| | - Joel S. Schuman
- Department of Ophthalmology and Visual Science Research Center University of Pittsburgh School of Medicine Pittsburgh PA USA
- Louis J. Fox Center for Vision Restoration of UPMC and the University of Pittsburgh Pittsburgh PA USA
- UPMC Eye Center Eye and Ear Institute Pittsburgh PA USA
- Department of Bioengineering Swanson School of Engineering University of Pittsburgh Pittsburgh PA USA
| | - Hongjun Liu
- Department of Microbiology and Molecular Genetics University of Pittsburgh School of Medicine Pittsburgh PA USA
- Department of Ophthalmology and Visual Science Research Center University of Pittsburgh School of Medicine Pittsburgh PA USA
- Louis J. Fox Center for Vision Restoration of UPMC and the University of Pittsburgh Pittsburgh PA USA
- UPMC Eye Center Eye and Ear Institute Pittsburgh PA USA
| |
Collapse
|
32
|
Wang X, Lin J, Arzeno A, Choi JY, Boccio J, Frieden E, Bhargava A, Maynard G, Tsai JC, Strittmatter SM. Intravitreal delivery of human NgR-Fc decoy protein regenerates axons after optic nerve crush and protects ganglion cells in glaucoma models. Invest Ophthalmol Vis Sci 2015; 56:1357-66. [PMID: 25655801 DOI: 10.1167/iovs.14-15472] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
PURPOSE Glaucoma is a major cause of vision loss due to retinal ganglion cell (RGC) degeneration. Therapeutic intervention controls increased IOP, but neuroprotection is unavailable. NogoReceptor1 (NgR1) limits adult central nervous system (CNS) axonal sprouting and regeneration. We examined NgR1 blocking decoy as a potential therapy by defining the pharmacokinetics of intravitreal NgR(310)-Fc, its promotion of RGC axonal regeneration following nerve crush, and its neuroprotective effect in a microbead glaucoma model. METHODS Human NgR1(310)-Fc was administered intravitreally, and levels were monitored in rat vitreal humor and retina. Axonal regeneration after optic nerve crush was assessed by cholera toxin β anterograde labeling. In a microbead model of glaucoma with increased IOP, the number of surviving and actively transporting RGCs was determined after 4 weeks by retrograde tracing with Fluro-Gold (FG) from the superior colliculus. RESULTS After intravitreal bolus administration, the terminal half-life of NgR1(310)-Fc between 1 and 7 days was approximately 24 hours. Injection of 5 μg protein once per week after optic nerve crush injury significantly increased RGCs with regenerating axons. Microbeads delivered to the anterior chamber increased pressure, and caused 15% reduction in FG-labeled RGCs of control rats, with a 40% reduction in large diameter RGCs. Intravitreal treatment with NgR1(310)-Fc did not reduce IOP, but maintained large diameter RGC density at control levels. CONCLUSIONS Human NgR1(310)-Fc has favorable pharmacokinetics in the vitreal space and rescues large diameter RGC counts from increased IOP. Thus, the NgR1 blocking decoy protein may have efficacy as a disease-modifying therapy for glaucoma.
Collapse
Affiliation(s)
- Xingxing Wang
- Program in Cellular Neuroscience, Neurodegeneration & Repair, Yale University School of Medicine, New Haven, Connecticut, United States Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, United States
| | - Jun Lin
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, Connecticut, United States
| | - Alexander Arzeno
- Program in Cellular Neuroscience, Neurodegeneration & Repair, Yale University School of Medicine, New Haven, Connecticut, United States Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, United States
| | - Jin Young Choi
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, Connecticut, United States
| | - Juliann Boccio
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, Connecticut, United States
| | - Eric Frieden
- Axerion Therapeutics, Branford, Connecticut, United States
| | - Ajay Bhargava
- Shakti BioResearch, Woodbridge, Connecticut, United States
| | - George Maynard
- Axerion Therapeutics, Branford, Connecticut, United States
| | - James C Tsai
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, Connecticut, United States
| | - Stephen M Strittmatter
- Program in Cellular Neuroscience, Neurodegeneration & Repair, Yale University School of Medicine, New Haven, Connecticut, United States Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, United States
| |
Collapse
|
33
|
Apara A, Goldberg JL. Molecular mechanisms of the suppression of axon regeneration by KLF transcription factors. Neural Regen Res 2014; 9:1418-21. [PMID: 25317150 PMCID: PMC4192940 DOI: 10.4103/1673-5374.139454] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2014] [Indexed: 01/11/2023] Open
Abstract
Molecular mechanisms of the Krüppel-like family of transcription factors (KLFs) have been studied more in proliferating cells than in post-mitotic cells such as neurons. We recently found that KLFs regulate intrinsic axon growth ability in central nervous system (CNS) neurons including retinal ganglion cells, and hippocampal and cortical neurons. With at least 15 of 17 KLF family members expressed in neurons and at least 5 structurally unique subfamilies, it is important to determine how this complex family functions in neurons to regulate the intricate genetic programs of axon growth and regeneration. By characterizing the molecular mechanisms of the KLF family in the nervous system, including binding partners and gene targets, and comparing them to defined mechanisms defined outside the nervous system, we may better understand how KLFs regulate neurite growth and axon regeneration.
Collapse
Affiliation(s)
| | - Jeffrey L Goldberg
- Shiley Eye Center, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
34
|
Curley JL, Catig GC, Horn-Ranney EL, Moore MJ. Sensory axon guidance with semaphorin 6A and nerve growth factor in a biomimetic choice point model. Biofabrication 2014; 6:035026. [PMID: 25189126 PMCID: PMC4170667 DOI: 10.1088/1758-5082/6/3/035026] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The direct effect of guidance cues on developing and regenerating axons in vivo is not fully understood, as the process involves a multiplicity of attractive and repulsive signals, presented both as soluble and membrane-bound ligands. A better understanding of axon guidance is critical to functional recovery following injury to the nervous system through improved outgrowth and mapping of damaged nerves. Due to their implications as inhibitors to central nervous system regeneration, we investigated the repulsive properties of semaphorin 6A and ephrin-B3 on E15 rat dorsal root ganglion explants, as well as possible interactions with soluble gradients of chemoattractive nerve growth factor (NGF). We employed a 3D biomimetic in vitro choice point model, which enabled the simple and rapid preparation of patterned gel growth matrices with quantifiable presentation of guidance cues in a specifiable manner that resembles the in vivo presentation of soluble and/or immobilized ligands. Neurites demonstrated an inhibitory response to immobilized Sema6A by lumbosacral dorsal root ganglion explants, while no such repulsion was observed for immobilized ephrin-B3 by explants at any spinal level. Interestingly, Sema6A inhibition could be partially attenuated in a concentration-dependent manner through the simultaneous presentation of soluble NGF gradients. The in vitro model described herein represents a versatile and valuable investigative tool in the quest for understanding developmental processes and improving regeneration following nervous system injury.
Collapse
Affiliation(s)
- J. Lowry Curley
- Lindy Boggs Bldg., Suite 500, Department of Biomedical Engineering, Tulane University, New Orleans, LA 70118, USA
| | - Gary C. Catig
- Lindy Boggs Bldg., Suite 500, Department of Biomedical Engineering, Tulane University, New Orleans, LA 70118, USA
| | - Elaine L. Horn-Ranney
- Lindy Boggs Bldg., Suite 500, Department of Biomedical Engineering, Tulane University, New Orleans, LA 70118, USA
| | - Michael J. Moore
- Lindy Boggs Bldg., Suite 500, Department of Biomedical Engineering, Tulane University, New Orleans, LA 70118, USA
| |
Collapse
|
35
|
Electrical stimulation of embryonic neurons for 1 hour improves axon regeneration and the number of reinnervated muscles that function. J Neuropathol Exp Neurol 2013; 72:697-707. [PMID: 23771218 DOI: 10.1097/nen.0b013e318299d376] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Motoneuron death after spinal cord injury or disease results in muscle denervation, atrophy, and paralysis. We have previously transplanted embryonic ventral spinal cord cells into the peripheral nerve to reinnervate denervated muscles and to reduce muscle atrophy, but reinnervation was incomplete. Here, our aim was to determine whether brief electrical stimulation of embryonic neurons in the peripheralnerve changes motoneuron survival, axon regeneration, and muscle reinnervation and function because neural depolarization is crucial for embryonic neuron survival and may promote activity-dependent axon growth. At 1 week after denervation by sciatic nerve section, embryonic day 14 to 15 cells were purified for motoneurons, injected into the tibial nerve of adult Fischer rats, and stimulated immediatelyfor up to 1 hour. More myelinated axons were present in tibial nerves 10 weeks after transplantation when transplants had been stimulated acutely at 1 Hz for 1 hour. More muscles were reinnervated if the stimulation treatment lasted for 1 hour. Reinnervation reduced muscle atrophy, with or without the stimulation treatment. These data suggest that brief stimulation of embryonic neurons promotes axon growth, which has a long-term impact on muscle reinnervation and function. Muscle reinnervation is important because it may enable the use of functional electrical stimulation to restore limb movements.
Collapse
|
36
|
Gao H, Zhang HL, Shou J, Chen L, Shen Y, Tang Q, Huang J, Zhu J. Towards retinal ganglion cell regeneration. Regen Med 2013; 7:865-75. [PMID: 23164085 DOI: 10.2217/rme.12.97] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Traumatic optic nerve injury and glaucoma are among the leading causes of incurable vision loss across the world. What is worse, neither pharmacological nor surgical interventions are significantly effective in reversing or halting the progression of vision loss. Advances in cell biology offer some hope for the victims of optic nerve damage and subsequent partial or complete visual loss. Retinal ganglion cells (RGCs) travel through the optic nerve and carry all visual signals to the brain. After injury, RGC axons usually fail to regrow and die, leading to irreversible loss of vision. Various kinds of cells and factors possess the ability to support the process of axon regeneration for RGCs. This article summarizes the latest advances in RGC regeneration.
Collapse
Affiliation(s)
- Huasong Gao
- Department of Neurosurgery, Fudan University Huashan Hospital, National Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Calkins DJ. Critical pathogenic events underlying progression of neurodegeneration in glaucoma. Prog Retin Eye Res 2012; 31:702-19. [PMID: 22871543 DOI: 10.1016/j.preteyeres.2012.07.001] [Citation(s) in RCA: 230] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2012] [Revised: 07/16/2012] [Accepted: 07/18/2012] [Indexed: 01/03/2023]
Abstract
Glaucoma is a common optic neuropathy with a complex etiology often linked to sensitivity to intraocular pressure. Though the precise mechanisms that mediate or transduce this sensitivity are not clear, the axon of the retinal ganglion cell appears to be vulnerable to disease-relevant stressors early in progression. One reason may be because the axon is generally thin for both its unmyelinated and myelinated segment and much longer than the thicker unmyelinated axons of other excitatory retinal neurons. This difference may predispose the axon to metabolic and oxidative injury, especially at distal sites where pre-synaptic terminals form connections in the brain. This idea is consistent with observations of early loss of anterograde transport at central targets and other signs of distal axonopathy that accompany physiological indicators of progression. Outright degeneration of the optic projection ensues after a critical period and, at least in animal models, is highly sensitive to cumulative exposure to elevated pressure in the eye. Stress emanating from the optic nerve head can induce not only distal axonopathy with aspects of dying back neuropathy, but also Wallerian degeneration of the optic nerve and tract and a proximal program involving synaptic and dendritic pruning in the retina. Balance between progressive and acute mechanisms likely varies with the level of stress placed on the unmyelinated axon as it traverses the nerve head, with more acute insult pushing the system toward quicker disassembly. A constellation of signaling factors likely contribute to the transduction of stress to the axon, so that degenerative events along the length of the optic projection progress in retinotopic fashion. This pattern leads to well-defined sectors of functional depletion, even at distal-most sites in the pathway. While ganglion cell somatic drop-out is later in progression, some evidence suggests that synaptic and dendritic pruning in the retina may be a more dynamic process. Structural persistence both in the retina and in central projection sites offers the possibility that intrinsic self-repair pathways counter pathogenic mechanisms to delay as long as possible outright loss of tissue.
Collapse
Affiliation(s)
- David J Calkins
- Department of Ophthalmology and Visual Sciences, The Vanderbilt Eye Institute, Vanderbilt University School of Medicine, 11435 MRB IV, 2215B Garland Avenue, Nashville, TN 37232, USA.
| |
Collapse
|
38
|
Abstract
Research into treatments for diseases of the CNS has made impressive strides in the past few decades, but therapeutic options are limited for many patients with CNS disorders. Nanotechnology has emerged as an exciting and promising new means of treating neurological disease, with the potential to fundamentally change the way we approach CNS-targeted therapeutics. Molecules can be nanoengineered to cross the blood-brain barrier, target specific cell or signalling systems, respond to endogenous stimuli, or act as vehicles for gene delivery, or as a matrix to promote axon elongation and support cell survival. The wide variety of available nanotechnologies allows the selection of a nanoscale material with the characteristics best suited to the therapeutic challenges posed by an individual CNS disorder. In this Review, we describe recent advances in the development of nanotechnology for the treatment of neurological disorders-in particular, neurodegenerative disease and malignant brain tumours-and for the promotion of neuroregeneration.
Collapse
Affiliation(s)
- Maya Srikanth
- Department of Neurology, Northwestern University Feinberg School of Medicine, Ward 10-233, 303 E. Chicago Avenue, Chicago, IL 60611, USA. maya@ fsm.northwestern.edu
| | | |
Collapse
|
39
|
Chang EE, Goldberg JL. Glaucoma 2.0: neuroprotection, neuroregeneration, neuroenhancement. Ophthalmology 2012; 119:979-86. [PMID: 22349567 DOI: 10.1016/j.ophtha.2011.11.003] [Citation(s) in RCA: 224] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Revised: 11/02/2011] [Accepted: 11/02/2011] [Indexed: 01/15/2023] Open
Abstract
Glaucoma is a progressive neurodegenerative disease of retinal ganglion cells (RGCs) associated with characteristic axon degeneration in the optic nerve. Clinically, our only method of slowing glaucomatous loss of vision is to reduce intraocular pressure (IOP), but lowering IOP is only partially effective and does not address the underlying susceptibility of RGCs to degeneration. We review the recent steps forward in our understanding of the pathophysiology of glaucoma and discuss how this understanding has given us a next generation of therapeutic targets by which to maintain RGC survival, protect or rebuild RGC connections in the retina and brain, and enhance RGC function.
Collapse
Affiliation(s)
- Elma E Chang
- University of Miami Miller School of Medicine, Miami, FL, USA
| | | |
Collapse
|
40
|
|
41
|
Moore DL, Apara A, Goldberg JL. Krüppel-like transcription factors in the nervous system: novel players in neurite outgrowth and axon regeneration. Mol Cell Neurosci 2011; 47:233-43. [PMID: 21635952 DOI: 10.1016/j.mcn.2011.05.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Accepted: 05/16/2011] [Indexed: 01/25/2023] Open
Abstract
The Krüppel-like family of transcription factors (KLFs) have been widely studied in proliferating cells, though very little is known about their role in post-mitotic cells, such as neurons. We have recently found that the KLFs play a role in regulating intrinsic axon growth ability in retinal ganglion cells (RGCs), a type of central nervous system (CNS) neuron. Previous KLF studies in other cell types suggest that there may be cell-type specific KLF expression patterns, and that their relative expression allows them to compete for binding sites, or to act redundantly to compensate for another's function. With at least 15 of 17 KLF family members expressed in neurons, it will be important for us to determine how this complex family functions to regulate the intricate gene programs of axon growth and regeneration. By further characterizing the mechanisms of the KLF family in the nervous system, we may better understand how they regulate neurite growth and axon regeneration.
Collapse
Affiliation(s)
- Darcie L Moore
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | | | | |
Collapse
|
42
|
Preserving and restoring optic nerve function. J Neuroophthalmol 2010; 30:303-4. [PMID: 21107120 DOI: 10.1097/wno.0b013e318200de91] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|