51
|
Abstract
Glaucoma is the second leading cause of blindness worldwide. Even though significant advances have been made in its management, currently available antiglaucoma therapies suffer from considerable drawbacks. Typically, the success and efficacy of glaucoma medications are undermined by their limited bioavailability to target tissues and the inadequate adherence demonstrated by patients with glaucoma. The latter is due to a gradual decrease in tolerability of lifelong topical therapies and the significant burden to patients of prescribed stepwise antiglaucoma regimens with frequent dosing which impact quality of life. On the other hand, glaucoma surgery is restricted by the inability of antifibrotic agents to efficiently control the wound healing process without causing severe collateral damage and long-term complications. Evolution of the treatment paradigm for patients with glaucoma will ideally include prevention of retinal ganglion cell degeneration by the successful delivery of neurotrophic factors, anti-inflammatory drugs, and gene therapies. Nanotechnology-based treatments may surpass the limitations of currently available glaucoma therapies through optimized targeted drug delivery, increased bioavailability, and controlled release. This review addresses the recent advances in glaucoma treatment strategies employing nanotechnology, including medical and surgical management, neuroregeneration, and neuroprotection.
Collapse
|
52
|
Therapeutic Potential of Mesenchymal Stem Cells and Their Secretome in the Treatment of Glaucoma. Stem Cells Int 2019; 2019:7869130. [PMID: 31949441 PMCID: PMC6948292 DOI: 10.1155/2019/7869130] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/09/2019] [Indexed: 12/15/2022] Open
Abstract
Glaucoma represents a group of progressive optic neuropathies characterized by gradual loss of retinal ganglion cells (RGCs), the neurons that conduct visual information from the retina to the brain. Elevated intraocular pressure (IOP) is considered the main reason for enhanced apoptosis of RGCs in glaucoma. Currently used therapeutic agents are not able to repopulate and/or regenerate injured RGCs and, therefore, are ineffective in most patients with advanced glaucoma. Accordingly, several new therapeutic approaches, including stem cell-based therapy, have been explored for the glaucoma treatment. In this review article, we emphasized current knowledge regarding molecular and cellular mechanisms responsible for beneficial effects of mesenchymal stem cells (MSCs) and their secretome in the treatment of glaucoma. MSCs produce neurotrophins and in an exosome-dependent manner supply injured RGCs with growth factors enhancing their survival and regeneration. Additionally, MSCs are able to generate functional RGC-like cells and induce proliferation of retinal stem cells. By supporting integrity of trabecular meshwork, transplanted MSCs alleviate IOP resulting in reduced loss of RGCs. Moreover, MSCs are able to attenuate T cell-driven retinal inflammation providing protection to the injured retinal tissue. In summing up, due to their capacity for neuroprotection and immunomodulation, MSCs and their secretome could be explored in upcoming clinical studies as new therapeutic agents for glaucoma treatment.
Collapse
|
53
|
Therapeutic Strategies for Attenuation of Retinal Ganglion Cell Injury in Optic Neuropathies: Concepts in Translational Research and Therapeutic Implications. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8397521. [PMID: 31828134 PMCID: PMC6885158 DOI: 10.1155/2019/8397521] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 10/07/2019] [Accepted: 10/28/2019] [Indexed: 12/19/2022]
Abstract
Retinal ganglion cell (RGC) death is the central and irreversible endpoint of optic neuropathies. Current management of optic neuropathies and glaucoma focuses on intraocular pressure-lowering treatment which is insufficient. As such, patients are effectively condemned to irreversible visual impairment. This review summarizes experimental treatments targeting RGCs over the last decade. In particular, we examine the various treatment modalities and determine their viability and limitations in translation to clinical practice. Experimental RGC treatment can be divided into (1) cell replacement therapy, (2) neuroprotection, and (3) gene therapy. For cell replacement therapy, difficulties remain in successfully integrating transplanted RGCs from various sources into the complex neural network of the human retina. However, there is significant potential for achieving full visual restoration with this technique. Neuroprotective strategies, in the form of pharmacological agents, nutritional supplementation, and neurotrophic factors, are viable strategies with encouraging results from preliminary noncomparative interventional case series. It is important to note, however, that most published studies are focused on glaucoma, with few treating optic neuropathies of other etiologies. Gene therapy, through the use of viral vectors, has shown promising results in clinical trials, particularly for diseases with specific genetic mutations like Leber's hereditary optic neuropathy. This treatment technique can be further extended to nonhereditary diseases, through transfer of genes promoting cell survival and neuroprotection. Crucially though, for gene therapy, teratogenicity remains a significant issue in translation to clinical practice.
Collapse
|
54
|
Normal tension glaucoma-like degeneration of the visual system in aged marmosets. Sci Rep 2019; 9:14852. [PMID: 31619716 PMCID: PMC6795850 DOI: 10.1038/s41598-019-51281-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 09/27/2019] [Indexed: 01/25/2023] Open
Abstract
The common marmoset (Callithrix jacchus) is a non-human primate that provides valuable models for neuroscience and aging research due to its anatomical similarities to humans and relatively short lifespan. This study was carried out to examine whether aged marmosets develop glaucoma, as seen in humans. We found that 11% of the aged marmosets presented with glaucoma-like characteristics; this incident rate is very similar to that in humans. Magnetic resonance imaging showed a significant volume loss in the visual cortex, and histological analyses confirmed the degeneration of the lateral geniculate nuclei and visual cortex in the affected marmosets. These marmosets did not have elevated intraocular pressure, but showed an increased oxidative stress level, low cerebrospinal fluid (CSF) pressure, and low brain-derived neurotrophic factor (BDNF) and TrkB expression in the retina, optic nerve head and CSF. Our findings suggest that marmosets have potential to provide useful information for the research of eye and the visual system.
Collapse
|
55
|
Gábriel R, Pöstyéni E, Dénes V. Neuroprotective Potential of Pituitary Adenylate Cyclase Activating Polypeptide in Retinal Degenerations of Metabolic Origin. Front Neurosci 2019; 13:1031. [PMID: 31649495 PMCID: PMC6794456 DOI: 10.3389/fnins.2019.01031] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 09/12/2019] [Indexed: 01/06/2023] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP1-38) is a highly conserved member of the secretin/glucagon/VIP family. The repressive effect of PACAP1-38 on the apoptotic machinery has been an area of active research conferring a significant neuroprotective potential onto this peptide. A remarkable number of studies suggest its importance in the etiology of neurodegenerative disorders, particularly in relation to retinal metabolic disorders. In our review, we provide short descriptions of various pathological conditions (diabetic retinopathy, excitotoxic retinal injury and ischemic retinal lesion) in which the remedial effect of PACAP has been well demonstrated in various animal models. Of all the pathological conditions, diabetic retinopathy seems to be the most intriguing as it develops in 75% of patients with type 1 and 50% of patients with type 2 diabetes, with concomitant progression to legal blindness in about 5%. Several animal models have been developed in recent years to study retinal degenerations and out of these glaucoma and age-related retina degeneration models bear human recapitulations. PACAP neuroprotection is thought to operate through enhanced cAMP production upon binding to PAC1-R. However, the underlying signaling network that leads to neuroprotection is not fully understood. We observed that (i) PACAP is not equally efficient in the above conditions; (ii) in some cases more than one signaling pathways are activated; (iii) the coupling of PAC1-R and signaling is stage dependent; and (iv) PAC1-R is not the only receptor that must be considered to interpret the effects in our experiments. These observations point to a complex signaling mechanism, that involves alternative routes besides the classical cAMP/protein kinase A pathway to evoke the outstanding neuroprotective action. Consequently, the possible contribution of the other two main receptors (VPAC1-R and VPAC2-R) will also be discussed. Finally, the potential medical use of PACAP in some retinal and ocular disorders will also be reviewed. By taking advantage of, low-cost synthesis technologies today, PACAP may serve as an alternative to the expensive treatment modelities currently available in ocular or retinal conditions.
Collapse
Affiliation(s)
- Robert Gábriel
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary.,János Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Etelka Pöstyéni
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary
| | - Viktória Dénes
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary
| |
Collapse
|
56
|
Kovacs K, Vaczy A, Fekete K, Kovari P, Atlasz T, Reglodi D, Gabriel R, Gallyas F, Sumegi B. PARP Inhibitor Protects Against Chronic Hypoxia/Reoxygenation-Induced Retinal Injury by Regulation of MAPKs, HIF1α, Nrf2, and NFκB. Invest Ophthalmol Vis Sci 2019; 60:1478-1490. [PMID: 30973576 DOI: 10.1167/iovs.18-25936] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose In the eye, chronic hypoxia/reoxygenation (H/R) contributes to the development of a number of ocular disorders. H/R induces the production of reactive oxygen species (ROS), leading to poly(ADP-ribose) polymerase-1 (PARP1) activation that promotes inflammation, cell death, and disease progression. Here, we analyzed the protective effects of the PARP1 inhibitor olaparib in H/R-induced retina injury and investigated the signaling mechanisms involved. Methods A rat retinal H/R model was used to detect histologic and biochemical changes in the retina. Results H/R induced reductions in the thickness of most retinal layers, which were prevented by olaparib. Furthermore, H/R caused increased levels of Akt and glycogen synthase kinase-3β phosphorylation, which were further increased by olaparib, contributing to retina protection. By contrast, H/R-induced c-Jun N-terminal kinase and p38 mitogen-activated protein kinases (MAPK) phosphorylation and activation were reduced by olaparib, via mitogen-activated protein kinase phosphatase 1 (MKP-1) expression. In addition, H/R-induced hypoxia-inducible factor 1α (HIF1α) levels were decreased by olaparib, which possibly contributed to reduced VEGF expression. Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) expression was slightly increased by H/R and was further activated by olaparib. Nuclear factor-κB (NFκB) was also activated by H/R through phosphorylation (Ser536) and acetylation (Lys310) of the p65 subunit, although this was significantly reduced by olaparib. Conclusions Olaparib reduced H/R-induced degenerative changes in retinal morphology. The protective mechanisms of olaparib most probably involved Nrf2 activation and ROS reduction, as well as normalization of HIF1α and related VEGF expression. In addition, olaparib reduced inflammation by NFκB dephosphorylation/inactivation, possibly via the PARP1 inhibition-MKP-1 activation-p38 MAPK inhibition pathway. PARP inhibitors represent potential therapeutics in H/R-induced retinal disease.
Collapse
Affiliation(s)
- Krisztina Kovacs
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, Pécs, Hungary
| | - Alexandra Vaczy
- Department of Anatomy, MTA-PTE PACAP Research Group, University of Pécs Medical School, Pécs, Hungary
| | - Katalin Fekete
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, Pécs, Hungary
| | - Petra Kovari
- Department of Anatomy, MTA-PTE PACAP Research Group, University of Pécs Medical School, Pécs, Hungary
| | - Tamas Atlasz
- Department of Anatomy, MTA-PTE PACAP Research Group, University of Pécs Medical School, Pécs, Hungary.,Department of Sportbiology, Faculty of Sciences, University of Pécs, Pécs, Hungary.,Szentagothai Research Centre, University of Pécs, Pécs, Hungary
| | - Dora Reglodi
- Department of Anatomy, MTA-PTE PACAP Research Group, University of Pécs Medical School, Pécs, Hungary
| | - Robert Gabriel
- Department of Biology, Faculty of Sciences, University of Pécs, Pécs, Hungary
| | - Ferenc Gallyas
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, Pécs, Hungary.,Szentagothai Research Centre, University of Pécs, Pécs, Hungary.,Nuclear-Mitochondrial Interactions Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Balazs Sumegi
- Department of Biochemistry and Medical Chemistry, University of Pécs Medical School, Pécs, Hungary.,Szentagothai Research Centre, University of Pécs, Pécs, Hungary.,Nuclear-Mitochondrial Interactions Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| |
Collapse
|
57
|
Khatib TZ, Martin KR. Neuroprotection in Glaucoma: Towards Clinical Trials and Precision Medicine. Curr Eye Res 2019; 45:327-338. [PMID: 31475591 DOI: 10.1080/02713683.2019.1663385] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Purpose: The eye is currently at the forefront of translational medicine and therapeutics. However, despite advances in technology, primary open-angle glaucoma remains the leading cause of irreversible blindness worldwide. Traditional intraocular pressure (IOP)-lowering therapies are often not sufficient to prevent progression to blindness, even in patients with access to high-quality healthcare. Neuroprotection strategies, which aim to boost the ability of target cells to withstand a pathological insult, have shown significant promise in animal models but none have shown clinically relevant efficacy in human clinical trials to date. We sought to evaluate the current status of neuroprotection clinical trials for glaucoma and identify limitations which have prevented translation of new glaucoma therapies to date.Methods: Literature searches identified English language references. Sources included MEDLINE, EMBASE, the Cochrane Library and Web of Science databases; reference lists of retrieved studies; and internet pages of relevant organisations, meetings and conference proceedings, and clinical trial registries.Results: We discuss six key neuroprotective strategies for glaucoma that have reached the clinical trial stage. Delivery of neurotrophic factors through gene therapy is also progressing towards glaucoma clinical trials. Refinements in trial design and the use of new modalities to define structural and functional endpoints may improve our assessment of disease activity and treatment efficacy. Advances in our understanding of compartmentalised glaucomatous degeneration and continued progress in the molecular profiling of glaucoma patients will enable us to predict individual risk and tailor treatment.Conclusion: New approaches to future glaucoma neuroprotection trials could improve the prospects for new glaucoma therapies. Glaucoma treatment tailored according to an individual's unique risk profile may become increasingly common in the future.
Collapse
Affiliation(s)
- Tasneem Z Khatib
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Eye Department, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.,Medical Sciences Division, University of Oxford, Oxford, UK
| | - Keith R Martin
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Eye Department, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.,Cambridge NIHR Biomedical Research Centre, Cambridge, UK.,Wellcome Trust - 5 MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.,Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Australia.,Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
| |
Collapse
|
58
|
Claes M, De Groef L, Moons L. Target-Derived Neurotrophic Factor Deprivation Puts Retinal Ganglion Cells on Death Row: Cold Hard Evidence and Caveats. Int J Mol Sci 2019; 20:E4314. [PMID: 31484425 PMCID: PMC6747494 DOI: 10.3390/ijms20174314] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 08/28/2019] [Accepted: 08/30/2019] [Indexed: 12/14/2022] Open
Abstract
Glaucoma and other optic neuropathies are characterized by axonal transport deficits. Axonal cargo travels back and forth between the soma and the axon terminus, a mechanism ensuring homeostasis and the viability of a neuron. An example of vital molecules in the axonal cargo are neurotrophic factors (NTFs). Hindered retrograde transport can cause a scarcity of those factors in the retina, which in turn can tilt the fate of retinal ganglion cells (RGCs) towards apoptosis. This postulation is one of the most widely recognized theories to explain RGC death in the disease progression of glaucoma and is known as the NTF deprivation theory. For several decades, research has been focused on the use of NTFs as a novel neuroprotective glaucoma treatment. Until now, results in animal models have been promising, but translation to the clinic has been highly disappointing. Are we lacking important knowledge to lever NTF therapies towards the therapeutic armamentarium? Or did we get the wrong end of the stick regarding the NTF deprivation theory? In this review, we will tackle the existing evidence and caveats advocating for and against the target-derived NTF deprivation theory in glaucoma, whilst digging into associated therapy efforts.
Collapse
Affiliation(s)
- Marie Claes
- Laboratory of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium
| | - Lies De Groef
- Laboratory of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium
| | - Lieve Moons
- Laboratory of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
59
|
Das M, Tang X, Mohapatra SS, Mohapatra S. Vision impairment after traumatic brain injury: present knowledge and future directions. Rev Neurosci 2019; 30:305-315. [PMID: 30226209 DOI: 10.1515/revneuro-2018-0015] [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: 02/15/2018] [Accepted: 06/15/2018] [Indexed: 01/23/2023]
Abstract
Traumatic brain injury (TBI) is a major cause of mortality and morbidity in the USA as well as in the world. As a result of TBI, the visual system is also affected often causing complete or partial visual loss, which in turn affects the quality of life. It may also lead to ocular motor dysfunction, defective accommodation, and impaired visual perception. As a part of the therapeutic strategy, early rehabilitative optometric intervention is important. Orthoptic therapy, medication, stem cell therapy, motor and attention trainings are the available treatment options. Gene therapy is one of the most promising emerging strategies. Use of state-of-the-art nanomedicine approaches to deliver drug(s) and/or gene(s) might enhance the therapeutic efficacy of the present and future modalities. More research is needed in these fields to improve the outcome of this debilitating condition. This review focuses on different visual pathologies caused by TBI, advances in pre-clinical and clinical research, and available treatment options.
Collapse
Affiliation(s)
- Mahasweta Das
- James A. Haley Veterans Administration Hospital, Tampa, FL 33612, USA.,Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL 33612, USA
| | - Xiaolan Tang
- James A. Haley Veterans Administration Hospital, Tampa, FL 33612, USA.,Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL 33612, USA
| | - Shyam S Mohapatra
- James A. Haley Veterans Administration Hospital, Tampa, FL 33612, USA.,Department of Internal Medicine, University of South Florida College of Medicine, Tampa, FL 33612, USA
| | - Subhra Mohapatra
- James A. Haley Veterans Administration Hospital, Tampa, FL 33612, USA.,Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL 33612, USA
| |
Collapse
|
60
|
Komáromy AM, Bras D, Esson DW, Fellman RL, Grozdanic SD, Kagemann L, Miller PE, Moroi SE, Plummer CE, Sapienza JS, Storey ES, Teixeira LB, Toris CB, Webb TR. The future of canine glaucoma therapy. Vet Ophthalmol 2019; 22:726-740. [PMID: 31106969 PMCID: PMC6744300 DOI: 10.1111/vop.12678] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 04/05/2019] [Accepted: 04/15/2019] [Indexed: 02/06/2023]
Abstract
Canine glaucoma is a group of disorders that are generally associated with increased intraocular pressure (IOP) resulting in a characteristic optic neuropathy. Glaucoma is a leading cause of irreversible vision loss in dogs and may be either primary or secondary. Despite the growing spectrum of medical and surgical therapies, there is no cure, and many affected dogs go blind. Often eyes are enucleated because of painfully high, uncontrollable IOP. While progressive vision loss due to primary glaucoma is considered preventable in some humans, this is mostly not true for dogs. There is an urgent need for more effective, affordable treatment options. Because newly developed glaucoma medications are emerging at a very slow rate and may not be effective in dogs, work toward improving surgical options may be the most rewarding approach in the near term. This Viewpoint Article summarizes the discussions and recommended research strategies of both a Think Tank and a Consortium focused on the development of more effective therapies for canine glaucoma; both were organized and funded by the American College of Veterinary Ophthalmologists Vision for Animals Foundation (ACVO-VAF). The recommendations consist of (a) better understanding of disease mechanisms, (b) early glaucoma diagnosis and disease staging, (c) optimization of IOP-lowering medical treatment, (d) new surgical therapies to control IOP, and (e) novel treatment strategies, such as gene and stem cell therapies, neuroprotection, and neuroregeneration. In order to address these needs, increases in research funding specifically focused on canine glaucoma are necessary.
Collapse
Affiliation(s)
- András M Komáromy
- College of Veterinary Medicine, Michigan State University, East Lansing, Michigan
| | - Dineli Bras
- Centro de Especialistas Veterinarios de Puerto Rico, San Juan, Puerto Rico
| | | | | | | | - Larry Kagemann
- U.S. Food and Drug Administration, Silver Spring, Maryland.,New York University School of Medicine, New York, New York.,Department of Ophthalmology, School of Medicine, University of Maryland, Baltimore, Maryland
| | - Paul E Miller
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Sayoko E Moroi
- Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan
| | - Caryn E Plummer
- College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | | | - Eric S Storey
- South Atlanta Veterinary Emergency & Specialty, Fayetteville, Georgia
| | - Leandro B Teixeira
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Carol B Toris
- Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, Nebraska
| | - Terah R Webb
- MedVet Medical & Cancer Centers for Pets, Worthington, Ohio
| |
Collapse
|
61
|
Application of CNTF or FGF-2 increases the number of M2-like macrophages after optic nerve injury in adult Rana pipiens. PLoS One 2019; 14:e0209733. [PMID: 31048836 PMCID: PMC6507305 DOI: 10.1371/journal.pone.0209733] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/17/2019] [Indexed: 02/07/2023] Open
Abstract
We have previously shown that a single application of the growth factors ciliary
neurotrophic factor (CNTF) or fibroblast growth factor 2 (FGF-2) to the crushed
optic nerve of the frog, Rana pipiens, increases the numbers
and elongation rate of regenerating retinal ganglion cell axons. Here we
investigate the effects of these factors on the numbers and types of macrophages
that invade the regeneration zone. In control PBS-treated nerves, many
macrophages are present 100 μm distal to the crush site at 1 week after injury;
their numbers halve by 2 weeks. A single application of CNTF at the time of
injury triples the numbers of macrophages at 1 week, with this increase compared
to control being maintained at 2 weeks. Application of FGF-2 is equally
effective at 1 week, but the macrophage numbers have fallen to control levels at
2 weeks. Immunostaining with a pan-macrophage marker, ED1, and a marker for
M2-like macrophages, Arg-1, showed that the proportion of the putative M2
phenotype remained at approximately 80% with all treatments. Electron microscopy
of the macrophages at 1 week shows strong phagocytic activity with all
treatments, with many vacuoles containing axon fragments and membrane debris. At
2 weeks with PBS or FGF-2 treatment the remaining macrophages are less
phagocytically active, containing mainly lipid inclusions. With CNTF treatment,
at 2 weeks many of the more numerous macrophages are still phagocytosing axonal
debris, although they also contain lipid inclusions. We conclude that the
increase in macrophage influx seen after growth factor application is beneficial
for the regenerating axons, probably due to more extensive removal of
degenerating distal axons, but also perhaps to secretion of growth-promoting
substances.
Collapse
|
62
|
Kim KI, Baek JY, Jeong JY, Nam JH, Park ES, Bok E, Shin WH, Chung YC, Jin BK. Delayed Treatment of Capsaicin Produces Partial Motor Recovery by Enhancing Dopamine Function in MPP +-lesioned Rats via Ciliary Neurotrophic Factor. Exp Neurobiol 2019; 28:289-299. [PMID: 31138996 PMCID: PMC6526113 DOI: 10.5607/en.2019.28.2.289] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 02/20/2019] [Accepted: 02/25/2019] [Indexed: 12/13/2022] Open
Abstract
Transient receptor potential vanilloid subtype 1 (TRPV1) on astrocytes prevents ongoing degeneration of nigrostriatal dopamine (DA) neurons in MPP+-lesioned rats via ciliary neurotrophic factor (CNTF). The present study determined whether such a beneficial effect of astrocytic TRPV1 could be achieved after completion of injury of DA neurons, rather than ongoing injury, which seems more relevant to therapeutics. To test this, the MPP+-lesioned rat model utilized here exhibited approximately 70~80% degeneration of nigrostriatal DA neurons that was completed at 2 weeks post medial forebrain bundle injection of MPP+. TRPV1 agonist, capsaicin (CAP), was intraperitoneally administered. CNTF receptor alpha neutralizing antibody (CNTFRαNAb) was nigral injected to evaluate the role of CNTF endogenously produced by astrocyte through TRPV1 activation on DA neurons. Delayed treatment of CAP produced a significant reduction in amphetamine-induced rotational asymmetry. Accompanying this behavioral recovery, CAP treatment increased CNTF levels and tyrosine hydroxylase (TH) activity in the substantia nigra pars compacta (SNpc), and levels of DA and its metabolites in the striatum compared to controls. Interestingly, behavioral recovery and increases in biochemical indices were not reflected in trophic changes of the DA system. Instead, behavioral recovery was temporal and dependent on the continuous presence of CAP treatment. The results suggest that delayed treatment of CAP increases nigral TH enzyme activity and striatal levels of DA and its metabolites by CNTF endogenously derived from CAP-activated astrocytes through TRPV1, leading to functional recovery. Consequently, these findings may be useful in the treatment of DA imbalances associated with Parkinson's disease.
Collapse
Affiliation(s)
- Kyoung In Kim
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Jeong Yeob Baek
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Jae Yeong Jeong
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Jin Han Nam
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Eun Su Park
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Eugene Bok
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Korea
| | - Won-Ho Shin
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Korea
| | - Young Cheul Chung
- Department of Biochemistry & Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Byung Kwan Jin
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul 02447, Korea.,Department of Biochemistry & Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| |
Collapse
|
63
|
Devoldere J, Peynshaert K, De Smedt SC, Remaut K. Müller cells as a target for retinal therapy. Drug Discov Today 2019; 24:1483-1498. [PMID: 30731239 DOI: 10.1016/j.drudis.2019.01.023] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/20/2018] [Accepted: 01/30/2019] [Indexed: 12/28/2022]
Abstract
Müller cells are specialized glial cells that span the entire retina from the vitreous cavity to the subretinal space. Their functional diversity and unique radial morphology render them particularly interesting targets for new therapeutic approaches. In this review, we reflect on various possibilities for selective Müller cell targeting and describe how some of their cellular mechanisms can be used for retinal neuroprotection. Intriguingly, cross-species investigation of their properties has revealed that Müller cells also have an essential role in retinal regeneration. Although many questions regarding this subject remain, it is clear that Müller cells have unique characteristics that make them suitable targets for the prevention and treatment of numerous retinal diseases.
Collapse
Affiliation(s)
- Joke Devoldere
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Karen Peynshaert
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Stefaan C De Smedt
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - Katrien Remaut
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium.
| |
Collapse
|
64
|
Ramlogan-Steel CA, Murali A, Andrzejewski S, Dhungel B, Steel JC, Layton CJ. Gene therapy and the adeno-associated virus in the treatment of genetic and acquired ophthalmic diseases in humans: Trials, future directions and safety considerations. Clin Exp Ophthalmol 2019; 47:521-536. [PMID: 30345694 DOI: 10.1111/ceo.13416] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 10/04/2018] [Accepted: 10/15/2018] [Indexed: 12/27/2022]
Abstract
Voretigene neparvovec-rzyl was recently approved for the treatment of Leber congenital amaurosis, and the use of gene therapy for eye disease is attracting even greater interest. The eye has immune privileged status, is easily accessible, requires a reduced dosage of therapy due to its size and is highly compartmentalized, significantly reducing systemic spread. Adeno-associated virus (AAV), with its low pathogenicity, prolonged expression profile and ability to transduce multiple cell types, has become the leading gene therapy vector. Target diseases have moved beyond currently untreatable inherited dystrophies to common, partially treatable acquired conditions such as exudative age-related macular degeneration and glaucoma, but use of the technology in these conditions imposes added obligations for caution in vector design. This review discusses the current status of AAV gene therapy trials in genetic and acquired ocular diseases, and explores new scientific developments, which could help ensure effective and safe use of the therapy in the future.
Collapse
Affiliation(s)
- Charmaine A Ramlogan-Steel
- LVF Ophthalmology Research Centre, Translational Research Institute, Brisbane, Australia.,Greenslopes Clinical School, Faculty of Medicine, University of Queensland, Greenslopes Hospital, Brisbane, Australia.,Medical and Applied Science, Central Queensland University, School of Health, Rockhampton, Australia
| | - Aparna Murali
- LVF Ophthalmology Research Centre, Translational Research Institute, Brisbane, Australia.,Greenslopes Clinical School, Faculty of Medicine, University of Queensland, Greenslopes Hospital, Brisbane, Australia
| | - Slawomir Andrzejewski
- LVF Ophthalmology Research Centre, Translational Research Institute, Brisbane, Australia.,Greenslopes Clinical School, Faculty of Medicine, University of Queensland, Greenslopes Hospital, Brisbane, Australia
| | - Bijay Dhungel
- Greenslopes Clinical School, Faculty of Medicine, University of Queensland, Greenslopes Hospital, Brisbane, Australia
| | - Jason C Steel
- Medical and Applied Science, Central Queensland University, School of Health, Rockhampton, Australia
| | - Christopher J Layton
- LVF Ophthalmology Research Centre, Translational Research Institute, Brisbane, Australia.,Greenslopes Clinical School, Faculty of Medicine, University of Queensland, Greenslopes Hospital, Brisbane, Australia
| |
Collapse
|
65
|
Adams CM, Stacy R, Rangaswamy N, Bigelow C, Grosskreutz CL, Prasanna G. Glaucoma - Next Generation Therapeutics: Impossible to Possible. Pharm Res 2018; 36:25. [PMID: 30547244 DOI: 10.1007/s11095-018-2557-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 11/12/2018] [Indexed: 12/28/2022]
Abstract
The future of next generation therapeutics for glaucoma is strong. The recent approval of two novel intraocular pressure (IOP)-lowering drugs with distinct mechanisms of action is the first in over 20 years. However, these are still being administered as topical drops. Efforts are underway to increase patient compliance and greater therapeutic benefits with the development of sustained delivery technologies. Furthermore, innovations from biologics- and gene therapy-based therapeutics are being developed in the context of disease modification, which are expected to lead to more permanent therapies for patients. Neuroprotection, including the preservation of retinal ganglion cells (RGCs) and optic nerve is another area that is actively being explored for therapeutic options. With improvements in imaging technologies and determination of new surrogate clinical endpoints, the therapeutic potential for translation of neuroprotectants is coming close to clinical realization. This review summarizes the aforementioned topics and other related aspects.
Collapse
Affiliation(s)
- Christopher M Adams
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research (NIBR),, Cambridge, Massachusetts, USA
| | - Rebecca Stacy
- Translational Medicine, Ophthalmology, NIBR, Cambridge, Massachusetts, USA
| | - Nalini Rangaswamy
- Ophthalmology Research, Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, Massachusetts, 02139, USA
| | - Chad Bigelow
- Ophthalmology Research, Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, Massachusetts, 02139, USA
| | - Cynthia L Grosskreutz
- Ophthalmology Research, Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, Massachusetts, 02139, USA
| | - Ganesh Prasanna
- Ophthalmology Research, Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, Massachusetts, 02139, USA.
| |
Collapse
|
66
|
Pietrucha-Dutczak M, Amadio M, Govoni S, Lewin-Kowalik J, Smedowski A. The Role of Endogenous Neuroprotective Mechanisms in the Prevention of Retinal Ganglion Cells Degeneration. Front Neurosci 2018; 12:834. [PMID: 30524222 PMCID: PMC6262299 DOI: 10.3389/fnins.2018.00834] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 10/25/2018] [Indexed: 12/14/2022] Open
Abstract
Retinal neurons are not able to undergo spontaneous regeneration in response to damage. A variety of stressors, i.e., UV radiation, high temperature, ischemia, allergens, and others, induce reactive oxygen species production, resulting in consecutive alteration of stress-response gene expression and finally can lead to cell apoptosis. Neurons have developed their own endogenous cellular protective systems. Some of them are preventing cell death and others are allowing functional recovery after injury. The high efficiency of these mechanisms is crucial for cell survival. In this review we focus on the contribution of the most recently studied endogenous neuroprotective factors involved in retinal ganglion cell (RGC) survival, among which, neurotrophic factors and their signaling pathways, processes regulating the redox status, and different pathways regulating cell death are the most important. Additionally, we summarize currently ongoing clinical trials for therapies for RGC degeneration and optic neuropathies, including glaucoma. Knowledge of the endogenous cellular protective mechanisms may help in the development of effective therapies and potential novel therapeutic targets in order to achieve progress in the treatment of retinal and optic nerve diseases.
Collapse
Affiliation(s)
- Marita Pietrucha-Dutczak
- Chair and Department of Physiology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Marialaura Amadio
- Department of Drug Sciences, Section of Pharmacology, University of Pavia, Pavia, Italy
| | - Stefano Govoni
- Department of Drug Sciences, Section of Pharmacology, University of Pavia, Pavia, Italy
| | - Joanna Lewin-Kowalik
- Chair and Department of Physiology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Adrian Smedowski
- Chair and Department of Physiology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
67
|
Flachsbarth K, Jankowiak W, Kruszewski K, Helbing S, Bartsch S, Bartsch U. Pronounced synergistic neuroprotective effect of GDNF and CNTF on axotomized retinal ganglion cells in the adult mouse. Exp Eye Res 2018; 176:258-265. [PMID: 30237104 DOI: 10.1016/j.exer.2018.09.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 09/04/2018] [Accepted: 09/16/2018] [Indexed: 01/09/2023]
Abstract
Neuroprotection is among the potential treatment options for glaucoma and other retinal pathologies characterized by the loss of retinal ganglion cells (RGCs). Here, we examined the impact of a neural stem (NS) cell-based intravitreal co-administration of two neuroprotective factors on the survival of axotomized RGCs. To this aim we used lentiviral vectors to establish clonal NS cell lines ectopically expressing either glial cell line-derived neurotrophic factor (GDNF) or ciliary neurotrophic factor (CNTF). The modified NS cell lines were intravitreally injected either separately or as a 1:1 mixture into adult mice one day after an optic nerve lesion, and the number of surviving RGCs was determined in retinal flat-mounts two, four and eight weeks after the lesion. For the transplantation experiments, we selected a GDNF- and a CNTF-expressing NS cell line that promoted the survival of axotomized RGCs with a similar efficacy. Eight weeks after the lesion, GDNF-treated retinas contained 3.8- and CNTF-treated retinas 3.7-fold more RGCs than control retinas. Of note, the number of surviving RGCs was markedly increased when both factors were administered simultaneously, with 14.3-fold more RGCs than in control retinas eight weeks after the lesion. GDNF and CNTF thus potently and synergistically rescued RGCs from axotomy-induced cell death, indicating that combinatorial neuroprotective approaches represent a promising strategy to effectively promote the survival of RGCs under pathological conditions.
Collapse
Affiliation(s)
- Kai Flachsbarth
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Wanda Jankowiak
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katharina Kruszewski
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sabine Helbing
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Susanne Bartsch
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Udo Bartsch
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
68
|
Laughter MR, Bardill JR, Ammar DA, Pena B, Calkins DJ, Park D. Injectable Neurotrophic Factor Delivery System Supporting Retinal Ganglion Cell Survival and Regeneration Following Optic Nerve Crush. ACS Biomater Sci Eng 2018; 4:3374-3383. [PMID: 31431919 PMCID: PMC6701853 DOI: 10.1021/acsbiomaterials.8b00803] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
In general, neurons belonging to the central nervous system (CNS), such as retinal ganglion cells (RGCs), do not regenerate. Due to this, strategies have emerged aimed at protecting and regenerating these cells. Neurotrophic factor (NTF) supplementation has been a promising approach but is limited by length of delivery and delivery vehicle. For this study, we tested a polymeric delivery system (sulfonated reverse thermal gel or SRTG) engineered to deliver cilliary neurotrophic factor (CNTF), while also being injectable. A rat optic nerve crush (ONC) model was used to determine the neuroprotective and regenerative capacity of our system. The results demonstrate that one single intravitreal injection of SRTG-CNTF following ONC showed significant protection of RGC survival at both 1 and 2 week time points, when compared to the control groups. Furthermore, there was no significant difference in the RGC count between the eyes that received the SRTG-CNTF following ONC and a healthy control eye. Intravitreal injection of the polymer system also induced noticeable axon regeneration 500 μm downstream from the lesion site compared to all other control groups. There was a significant increase in Müller cell response in groups that received the SRTG-CNTF injection following optic nerve crush also indicative of a regenerative response. Finally, higher concentrations of CNTF released from SRTG-CNTF showed a protective effect on RGCs and Müller cell response at a longer time point (4 weeks). In conclusion, we were able to show a neuroprotective and regenerative effect of this polymer SRTG-CNTF delivery system and the viability for treatment of neurodegenerations.
Collapse
Affiliation(s)
- Melissa R. Laughter
- Department of Bioengineering, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - James R. Bardill
- Department of Bioengineering, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - David A. Ammar
- Department of Ophthalmology, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Brisa Pena
- Department of Bioengineering, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - David J. Calkins
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Daewon Park
- Department of Bioengineering, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado 80045, United States
| |
Collapse
|
69
|
Eriksen AZ, Eliasen R, Oswald J, Kempen PJ, Melander F, Andresen TL, Young M, Baranov P, Urquhart AJ. Multifarious Biologic Loaded Liposomes that Stimulate the Mammalian Target of Rapamycin Signaling Pathway Show Retina Neuroprotection after Retina Damage. ACS NANO 2018; 12:7497-7508. [PMID: 30004669 PMCID: PMC6117751 DOI: 10.1021/acsnano.8b00596] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 07/13/2018] [Indexed: 05/08/2023]
Abstract
A common event in optic neuropathies is the loss of axons and death of retinal ganglion cells (RGCs) resulting in irreversible blindness. Mammalian target of rapamycin (mTOR) signaling pathway agonists have been shown to foster axon regeneration and RGC survival in animal models of optic nerve damage. However, many challenges remain in developing therapies that exploit cell growth and tissue remodeling including (i) activating/inhibiting cell pathways synergistically, (ii) avoiding tumorigenesis, and (iii) ensuring appropriate physiological tissue function. These challenges are further exacerbated by the need to overcome ocular physiological barriers and clearance mechanisms. Here we present liposomes loaded with multiple mTOR pathway stimulating biologics designed to enhance neuroprotection after retina damage. Liposomes were loaded with ciliary neurotrophic factor, insulin-like growth factor 1, a lipopeptide N-fragment osteopontin mimic, and lipopeptide phosphatase tension homologue inhibitors for either the ATP domain or the c-terminal tail. In a mouse model of N-methyl-d-aspartic acid induced RGC death, a single intravitreal administration of liposomes reduced both RGC death and loss of retina electrophysiological function. Furthermore, combining liposomes with transplantation of induced pluripotent stem cell derived RGCs led to an improved electrophysiological outcome in mice. The results presented here show that liposomes carrying multiple signaling pathway modulators can facilitate neuroprotection and transplant electrophysiological outcome.
Collapse
Affiliation(s)
- Anne Z. Eriksen
- DTU
Nanotech, Department of Micro- and Nanotechnology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Rasmus Eliasen
- DTU
Nanotech, Department of Micro- and Nanotechnology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Julia Oswald
- Schepens
Eye Research Institute, Massachusetts Eye and Ear, 20 Staniford Street, Boston, Massachusetts 02114, United States
| | - Paul J. Kempen
- DTU
Nanotech, Department of Micro- and Nanotechnology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Fredrik Melander
- DTU
Nanotech, Department of Micro- and Nanotechnology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Thomas L. Andresen
- DTU
Nanotech, Department of Micro- and Nanotechnology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Michael Young
- Schepens
Eye Research Institute, Massachusetts Eye and Ear, 20 Staniford Street, Boston, Massachusetts 02114, United States
| | - Petr Baranov
- Schepens
Eye Research Institute, Massachusetts Eye and Ear, 20 Staniford Street, Boston, Massachusetts 02114, United States
| | - Andrew J. Urquhart
- DTU
Nanotech, Department of Micro- and Nanotechnology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| |
Collapse
|
70
|
Guymer C, Wood JPM, Chidlow G, Casson RJ. Neuroprotection in glaucoma: recent advances and clinical translation. Clin Exp Ophthalmol 2018; 47:88-105. [DOI: 10.1111/ceo.13336] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 05/21/2018] [Accepted: 06/06/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Chelsea Guymer
- Ophthalmic Research Laboratory, South Australian Institute of Ophthalmology; University of Adelaide; Adelaide South Australia Australia
| | - John PM Wood
- Ophthalmic Research Laboratory, South Australian Institute of Ophthalmology; University of Adelaide; Adelaide South Australia Australia
| | - Glyn Chidlow
- Ophthalmic Research Laboratory, South Australian Institute of Ophthalmology; University of Adelaide; Adelaide South Australia Australia
| | - Robert J Casson
- Ophthalmic Research Laboratory, South Australian Institute of Ophthalmology; University of Adelaide; Adelaide South Australia Australia
| |
Collapse
|
71
|
Wang WJ, Jin W, Yang AH, Chen Z, Xing YQ. Protective effects of ciliary neurotrophic factor on the retinal ganglion cells by injure of hydrogen peroxide. Int J Ophthalmol 2018; 11:923-928. [PMID: 29977802 DOI: 10.18240/ijo.2018.06.05] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 11/22/2017] [Indexed: 12/11/2022] Open
Abstract
AIM To explore the effect of ciliary neurotrophic factor (CNTF) on retinal ganglion cell (RGC)-5 induced by hydrogen peroxide (H2O2). METHODS After cell adherence, RGC-5 culture medium was changed to contain different concentrations of H2O2 from 50 to 150 µmol/L at four time points (0.5, 1, 1.5 and 2h) to select the concentration and time point for H2O2 induced model. Two different ways of interventions for injured RGC-5 cells respectively were CNTF as an addition in the culture medium or recombinant lentiviral plasmid carrying CNTF gene transfecting bone mesenchymal stem cells (BMSCs) for co-culture with RGC-5. RESULTS Compared to the control group, H2O2 led to RGC-5 death closely associated with concentrations and action time of H2O2 and we chose 125 µmol/L and 2h to establish the H2O2-induced model. While CNTF inhibited the loss of RGC-5 cells obviously with a dose-dependent survival rate. Nevertheless two administration routes had different survival rate yet higher rate in recombinant lentiviral plasmid group but there were no statistically significant differences. CONCLUSION Both the two administration routes of CNTF have effects on RGC-5 cells induced by H2O2. If their own advantages were combined, there may be a better administration route.
Collapse
Affiliation(s)
- Wen-Jun Wang
- Eye Center, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Wei Jin
- Eye Center, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - An-Huai Yang
- Eye Center, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Zhen Chen
- Eye Center, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Yi-Qiao Xing
- Eye Center, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| |
Collapse
|
72
|
Ma M, Xu Y, Xiong S, Zhang J, Gu Q, Ke B, Xu X. Involvement of ciliary neurotrophic factor in early diabetic retinal neuropathy in streptozotocin-induced diabetic rats. Eye (Lond) 2018; 32:1463-1471. [PMID: 29795129 PMCID: PMC6137181 DOI: 10.1038/s41433-018-0110-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 03/06/2018] [Accepted: 03/18/2018] [Indexed: 12/24/2022] Open
Abstract
Objective Ciliary neurotrophic factor (CNTF) has been evaluated as a candidate therapeutic agent for diabetes and its neural complications. However, its role in diabetic retinopathy has not been fully elucidated. Methods This is a randomized unblinded animal experiment. Wistar rats with streptozocin (STZ)-induced diabetes were regularly injected with CNTF or vehicle control in their vitreous bodies beginning at 2 weeks after STZ injection. A total of five injections were used. In diabetic rats, the levels of CNTF and neurotrophin-3 (NT-3) were evaluated by enzyme-linked immunosorbent assays (ELISA) and real-time PCR. The abundance of tyrosine hydroxylase (TH) and β-III tubulin was detected by western blot. Transferase-mediated dUTP nick-end labeling staining (TUNEL) was used to detect cell apoptosis in the retinal tissue. The activation of caspase-3 was also measured. Results The protein and mRNA levels of CNTF in diabetic rat retinas were reduced compared to control rats. In addition, retinal ganglion cells (RGCs) and dopaminergic amacrine cells appeared to undergo degeneration in diabetic rat retinas, as revealed by transferase-mediated dUTP nick-end labeling staining (TUNEL). Tyrosine hydroxylase (TH) and β-III tubulin protein levels also decreased significantly. Intraocular administration of CNTF rescued RGCs and dopaminergic amacrine cells from neurodegeneration and counteracted the downregulation of β-III tubulin and TH expression, thus demonstrating its therapeutic potential. Conclusion Our study suggests that early diabetic retinal neuropathy involves the reduced expression of CNTF and can be ameliorated by an exogenous supply of this neurotrophin.
Collapse
Affiliation(s)
- Mingming Ma
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.,Department of Ophthalmology, Shanghai General Hospital, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Yupeng Xu
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.,Department of Ophthalmology, Shanghai General Hospital, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Shuyu Xiong
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.,Department of Ophthalmology, Shanghai General Hospital, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Jian Zhang
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.,Department of Ophthalmology, Shanghai General Hospital, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Qing Gu
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.,Department of Ophthalmology, Shanghai General Hospital, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Bilian Ke
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.,Department of Ophthalmology, Shanghai General Hospital, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Xun Xu
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China. .,Department of Ophthalmology, Shanghai General Hospital, Shanghai, China. .,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.
| |
Collapse
|
73
|
Progress in Gene Therapy to Prevent Retinal Ganglion Cell Loss in Glaucoma and Leber's Hereditary Optic Neuropathy. Neural Plast 2018; 2018:7108948. [PMID: 29853847 PMCID: PMC5954906 DOI: 10.1155/2018/7108948] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/04/2018] [Indexed: 12/24/2022] Open
Abstract
The eye is at the forefront of the application of gene therapy techniques to medicine. In the United States, a gene therapy treatment for Leber's congenital amaurosis, a rare inherited retinal disease, recently became the first gene therapy to be approved by the FDA for the treatment of disease caused by mutations in a specific gene. Phase III clinical trials of gene therapy for other single-gene defect diseases of the retina and optic nerve are also currently underway. However, for optic nerve diseases not caused by single-gene defects, gene therapy strategies are likely to focus on slowing or preventing neuronal death through the expression of neuroprotective agents. In addition to these strategies, there has also been recent interest in the potential use of precise genome editing techniques to treat ocular disease. This review focuses on recent developments in gene therapy techniques for the treatment of glaucoma and Leber's hereditary optic neuropathy (LHON). We discuss recent successes in clinical trials for the treatment of LHON using gene supplementation therapy, promising neuroprotective strategies that have been employed in animal models of glaucoma and the potential use of genome editing techniques in treating optic nerve disease.
Collapse
|
74
|
Schultz R, Krug M, Precht M, Wohl SG, Witte OW, Schmeer C. Frataxin overexpression in Müller cells protects retinal ganglion cells in a mouse model of ischemia/reperfusion injury in vivo. Sci Rep 2018; 8:4846. [PMID: 29555919 PMCID: PMC5859167 DOI: 10.1038/s41598-018-22887-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 03/02/2018] [Indexed: 01/28/2023] Open
Abstract
Müller cells are critical for retinal function and neuronal survival but can become detrimental in response to retinal ischemia and increased oxidative stress. Elevated oxidative stress increases expression of the mitochondrial enzyme frataxin in the retina, and its overexpression is neuroprotective after ischemia. Whether frataxin expression in Müller cells might improve their function and protect neurons after ischemia is unknown. The aim of this study was to evaluate the effect of frataxin overexpression in Müller cells on neuronal survival after retinal ischemia/reperfusion in the mouse in vivo. Retinal ischemia/reperfusion was induced in mice overexpressing frataxin in Müller cells by transient elevation of intraocular pressure. Retinal ganglion cells survival was determined 14 days after lesion. Expression of frataxin, antioxidant enzymes, growth factors and inflammation markers was determined with qRT-PCR, Western blotting and immunohistochemistry 24 hours after lesion. Following lesion, there was a 65% increase in the number of surviving RGCs in frataxin overexpressing mice. Improved survival was associated with increased expression of the antioxidant enzymes Gpx1 and Sod1 as well as the growth factors Cntf and Lif. Additionally, microglial activation was decreased in these mice. Therefore, support of Müller cell function constitutes a feasible approach to reduce neuronal degeneration after ischemia.
Collapse
Affiliation(s)
- Rowena Schultz
- Department of Ophthalmology, Jena University Hospital, Jena, Germany
| | - Melanie Krug
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Michel Precht
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Stefanie G Wohl
- Department of Biological Structure, University of Washington Seattle, Seattle, United States
| | - Otto W Witte
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Christian Schmeer
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany.
| |
Collapse
|
75
|
Graca AB, Hippert C, Pearson RA. Müller Glia Reactivity and Development of Gliosis in Response to Pathological Conditions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1074:303-308. [PMID: 29721957 DOI: 10.1007/978-3-319-75402-4_37] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Within the mammalian retina, both Müller glia and astrocytes display reactivity in response to many forms of retinal injury and disease in a process termed gliosis. Reactive gliosis is a complex process that is considered to represent a cellular response to protect the retina from further damage and to promote its repair following pathological insult. It includes morphological, biochemical and physiological changes, which may vary depending on the type and degree of the initial injury. Not only does gliosis have numerous triggers, but also there is a great degree of heterogeneity in the glial response, creating multiple levels of complexity. For these reasons, understanding the process of glial scar formation and how this process differs in different pathological conditions and finding strategies to circumvent these barriers represent major challenges to the advancement of many ocular therapies.
Collapse
Affiliation(s)
- Anna B Graca
- Department of Genetics, University College London Institute of Ophthalmology, London, UK.
| | - Claire Hippert
- Roche, Stem Cell Platform, Chemical Biology Roche Pharma Research and Early Development, Basel, Switzerland
| | - Rachael A Pearson
- Department of Genetics, University College London Institute of Ophthalmology, London, UK.
| |
Collapse
|
76
|
|
77
|
Chitranshi N, Dheer Y, Abbasi M, You Y, Graham SL, Gupta V. Glaucoma Pathogenesis and Neurotrophins: Focus on the Molecular and Genetic Basis for Therapeutic Prospects. Curr Neuropharmacol 2018; 16:1018-1035. [PMID: 29676228 PMCID: PMC6120108 DOI: 10.2174/1570159x16666180419121247] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 04/10/2018] [Accepted: 04/18/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Retinal ganglion cell (RGC) degeneration is a major feature of glaucoma pathology. Neuroprotective approaches that delay or halt the progression of RGC loss are needed to prevent vision loss which can occur even after conventional medical or surgical treatments to lower intraocular pressure. OBJECTIVE The aim of this review was to examine the progress in genetics and cellular mechanisms associated with endoplasmic reticulum (ER) stress, RGC dysfunction and cell death pathways in glaucoma. MATERIALS AND METHODS Here, we review the involvement of neurotrophins like brain derived neurotrophic factor (BDNF) and its high affinity receptor tropomyosin receptor kinase (TrkB) in glaucoma. The role of ER stress markers in human and animal retinas in health and disease conditions is also discussed. Further, we analysed the literature highlighting genetic linkage in the context of primary open angle glaucoma and suggested mechanistic insights into potential therapeutic options relevant to glaucoma management. RESULTS The literature review of the neurobiology underlying neurotrophin pathways, ER stress and gene associations provide critical insights into association of RGCs death in glaucoma. Alteration in signalling pathway is associated with increased risk of misfolded protein aggregation in ER promoting RGC apoptosis. Several genes that are linked with neurotrophin signalling pathways have been reported to be associated with glaucoma pathology. CONCLUSION Understanding genetic heterogeneity and involvement of neurotrophin biology in glaucoma could help to understand the complex pathophysiology of glaucoma. Identification of novel molecular targets will be critical for drug development and provide neuroprotection to the RGCs and optic nerve.
Collapse
Affiliation(s)
- Nitin Chitranshi
- Address correspondence to this author at the Faculty of Medicine and Health Sciences, 75, Talavera Road, Macquarie University, Sydney, NSW 2109, Australia; Tel: +61-298502760; E-mail:
| | | | | | | | | | | |
Collapse
|
78
|
Ciliary neurotrophic factor in patients with primary open-angle glaucoma and age-related cataract. Mol Vis 2017; 23:799-809. [PMID: 29225456 PMCID: PMC5710971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 11/15/2017] [Indexed: 11/17/2022] Open
Abstract
Purpose To study the ciliary neurotrophic factor (CNTF) concentration in the aqueous humor (AH), lacrimal fluid (LF), and blood serum (BS) in patients with age-related cataract and primary open-angle glaucoma (POAG). Methods CNTF concentrations were studied in 61 patients with age-related cataract, 55 patients with POAG combined with cataract, and 29 healthy controls (one eye in each person). Preliminary experiments permitted us to extend the minimum quantifiable value of the CNTF Quantikine enzyme-linked immunosorbent assay (ELISA) kit to 2.5 pg/ml. Results The levels of CNTF in LF and BS did not differ in patients with cataract and controls. The CNTF concentration (pg/ml) in patients with POAG and cataract was lower than in patients with cataract (p<0.001) in AH (39.9±26.2 versus 57.2±25.6) and in LF (25.7±14.9 versus 39.9±18.0). The differences were not statistically significant for the CNTF level in BS (5.45±4.72 versus 5.96±4.92) and the AH/LF ratio (1.69±1.05 versus 1.58±0.70). In the patients with POAG, the AH level of CNTF correlated with the visual field index (Pearson's correlation coefficient r = 0.35, p = 0.01). A statistically significant decrease in the AH and LF concentrations of CNTF was observed in patients in all stages of POAG compared with the cataract group. This decrease was particularly prominent in patients with severe glaucoma. Compared to patients with combined early and moderate stages of disease patients with advanced glaucoma showed an insignificant reduction in the median CNTF concentration in AH and LF. The serum CNTF concentration did not show any dependence on the glaucoma stage. The CNTF concentration in the AH strongly correlated with the CNTF concentration in the LF (r=0.71, p<0.000). A formula was suggested to calculate the concentration of CNTF in AH based on the CNTF concentration in LF. Conclusions The CNTF concentration is reduced in the AH and LF of patients with POAG, especially in those with severe visual field loss. The CNTF concentration in AH and LF showed a strong correlation, and this phenomenon opens up new options for a noninvasive estimation of the CNTF concentration in AH. The CNTF concentration established in the AH, LF, and BS of patients with age-related cataract can serve as normative data for persons older than 50 years old.
Collapse
|
79
|
Ghasemi M, Alizadeh E, Saei Arezoumand K, Fallahi Motlagh B, Zarghami N. Ciliary neurotrophic factor (CNTF) delivery to retina: an overview of current research advancements. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:1694-1707. [PMID: 29065723 DOI: 10.1080/21691401.2017.1391820] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The intraocular administration of the ciliary neurotrophic factor (CNTF) has been found to attenuate the photoreceptor degeneration and preserve retinal functions in the animal research models of the inherited or induced retinal disease. Studies with the aim of CNTF transfer to the posterior segment inside the eye have been directed to determine the best method for its administration. An ideal delivery method would overcome the eye drug elimination mechanisms or barriers and provide the sustained release of the CNTF into retina in the safest fashion with the minimum harm to the quality of life. This review focuses on the present state of CNTF delivery to retina, also provides an overview of available technologies and their challenges.
Collapse
Affiliation(s)
- Maryam Ghasemi
- a The Umbilical Cord Stem Cell Research Center (UCSRC) , Tabriz University of Medical Sciences , Tabriz , Iran.,b Department of Medical Biotechnology, Faculty of Advanced Medical Sciences , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Effat Alizadeh
- a The Umbilical Cord Stem Cell Research Center (UCSRC) , Tabriz University of Medical Sciences , Tabriz , Iran.,b Department of Medical Biotechnology, Faculty of Advanced Medical Sciences , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Khatereh Saei Arezoumand
- b Department of Medical Biotechnology, Faculty of Advanced Medical Sciences , Tabriz University of Medical Sciences , Tabriz , Iran
| | | | - Nosratollah Zarghami
- a The Umbilical Cord Stem Cell Research Center (UCSRC) , Tabriz University of Medical Sciences , Tabriz , Iran.,b Department of Medical Biotechnology, Faculty of Advanced Medical Sciences , Tabriz University of Medical Sciences , Tabriz , Iran.,d Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine , Tabriz University of Medical Sciences , Tabriz , Iran
| |
Collapse
|
80
|
Chidlow G, Ebneter A, Wood JPM, Casson RJ. Evidence Supporting an Association Between Expression of Major Histocompatibility Complex II by Microglia and Optic Nerve Degeneration During Experimental Glaucoma. J Glaucoma 2017; 25:681-91. [PMID: 27253969 DOI: 10.1097/ijg.0000000000000447] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
AIM We acquired age-matched and sex-matched Sprague-Dawley rats from 2 independent breeding establishments. Serendipitously, we observed that constitutive, and bacterial toxin-induced, expression of major histocompatibility complex (MHC) class II RT1B chain in the uveal tract was much lower in one of the cohorts. Activated microglia are known to upregulate MHC II RT1B expression during optic nerve (ON) degeneration induced by raised intraocular pressure (IOP). We investigated whether, in a model of experimental glaucoma, microglial upregulation of MHC II RT1B was less efficacious and ON degeneration correspondingly less severe in the cohort of rats with low MHC II RT1B expression. METHODS Experimental glaucoma was induced by lasering the trabecular meshwork using a standard protocol. After 2 weeks of elevated IOP, retinal ganglion cells (RGC) survival, ON degeneration, and microglial responses were determined in both cohorts of rats. RESULTS Raised IOP-induced expression of MHC II RT1B by microglia was muted in the "Low" cohort compared with the "High" cohort. Axonal degeneration, RGC loss, and microgliosis were all significantly lower in the cohort of rats with low basal and induced expression of MHC II RT1B, despite both cohorts displaying IOP responses that were indistinguishable in terms of peak IOP and IOP exposure. CONCLUSIONS Expression of MHC II RT1B by activated microglia in the ON during experimental glaucoma was associated with more severe RGC degeneration. Further studies are needed to elucidate the role of MHC II during experimental glaucoma.
Collapse
Affiliation(s)
- Glyn Chidlow
- *Ophthalmic Research Laboratories, South Australian Institute of Ophthalmology, Hanson Institute Centre for Neurological Diseases †Department of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, SA, Australia ‡Department of Ophthalmology, Bern University Hospital and University of Bern, Inselspital, Bern, Switzerland
| | | | | | | |
Collapse
|
81
|
Toft-Kehler AK, Skytt DM, Kolko M. A Perspective on the Müller Cell-Neuron Metabolic Partnership in the Inner Retina. Mol Neurobiol 2017; 55:5353-5361. [PMID: 28929338 DOI: 10.1007/s12035-017-0760-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 08/31/2017] [Indexed: 10/18/2022]
Abstract
The Müller cells represent the predominant macroglial cell in the retina. In recent decades, Müller cells have been acknowledged to be far more influential on neuronal homeostasis in the retina than previously assumed. With its unique localization, spanning the entire retina being interposed between the vessels and neurons, Müller cells are responsible for the functional and metabolic support of the surrounding neurons. As a consequence of major energy demands in the retina, high levels of glucose are consumed and processed by Müller cells. The present review provides a perspective on the symbiotic relationship between Müller cells and inner retinal neurons on a cellular level by emphasizing the essential role of energy metabolism within Müller cells in relation to retinal neuron survival.
Collapse
Affiliation(s)
- A K Toft-Kehler
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - D M Skytt
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Miriam Kolko
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark. .,Department of Ophthalmology, Zealand University Hospital, Vestermarksvej 23, 4000, Roskilde, Denmark. .,Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet, Nordre Ringvej 57, 2600, Glostrup, Denmark.
| |
Collapse
|
82
|
Gao FJ, Zhang SH, Xu P, Yang BQ, Zhang R, Cheng Y, Zhou XJ, Huang WJ, Wang M, Chen JY, Sun XH, Wu JH. Quercetin Declines Apoptosis, Ameliorates Mitochondrial Function and Improves Retinal Ganglion Cell Survival and Function in In Vivo Model of Glaucoma in Rat and Retinal Ganglion Cell Culture In Vitro. Front Mol Neurosci 2017; 10:285. [PMID: 28936163 PMCID: PMC5594060 DOI: 10.3389/fnmol.2017.00285] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 08/23/2017] [Indexed: 12/17/2022] Open
Abstract
Glaucoma is a progressive neuropathy characterized by the loss of retinal ganglion cells (RGCs). Strategies that delay or halt RGC loss have been recognized as potentially beneficial for rescuing vision in glaucoma patients. Quercetin (Qcn) is a natural and important dietary flavonoid compound, widely distributed in fruits and vegetables. Mounting evidence suggests that Qcn has numerous neuroprotective effects. However, whether Qcn exerts neuroprotective effects on RGC in glaucoma is poorly understood. In this study, we investigated the protective effect of Qcn against RGC damage in a rat chronic ocular hypertension (COHT) model invivo and hypoxia-induced primary cultured RGC damage in vitro, and we further explored the underlying neuroprotective mechanisms. We found that Qcn not only improved RGC survival and function from a very early stage of COHT invivo, it promoted the survival of hypoxia-treated primary cultured RGCs invitro via ameliorating mitochondrial function and preventing mitochondria-mediated apoptosis. Our findings suggest that Qcn has direct protective effects on RGCs that are independent of lowering the intraocular pressure (IOP). Qcn may be a promising therapeutic agent for improving RGC survival and function in glaucomatous neurodegeneration.
Collapse
Affiliation(s)
- Feng-Juan Gao
- Eye Institute, Eye and ENT Hospital, College of Medicine, Fudan UniversityShanghai, China
| | - Sheng-Hai Zhang
- Eye Institute, Eye and ENT Hospital, College of Medicine, Fudan UniversityShanghai, China.,State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan UniversityShanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality,Shanghai, China.,Key Laboratory of Myopia, Ministry of HealthShanghai, China
| | - Ping Xu
- Eye Institute, Eye and ENT Hospital, College of Medicine, Fudan UniversityShanghai, China.,State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan UniversityShanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality,Shanghai, China.,Key Laboratory of Myopia, Ministry of HealthShanghai, China
| | - Bo-Qi Yang
- Eye Institute, Eye and ENT Hospital, College of Medicine, Fudan UniversityShanghai, China.,State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan UniversityShanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality,Shanghai, China.,Key Laboratory of Myopia, Ministry of HealthShanghai, China
| | - Rong Zhang
- Eye Institute, Eye and ENT Hospital, College of Medicine, Fudan UniversityShanghai, China.,State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan UniversityShanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality,Shanghai, China.,Key Laboratory of Myopia, Ministry of HealthShanghai, China
| | - Yun Cheng
- Eye Institute, Eye and ENT Hospital, College of Medicine, Fudan UniversityShanghai, China.,State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan UniversityShanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality,Shanghai, China.,Key Laboratory of Myopia, Ministry of HealthShanghai, China
| | - Xu-Jiao Zhou
- Eye Institute, Eye and ENT Hospital, College of Medicine, Fudan UniversityShanghai, China.,State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan UniversityShanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality,Shanghai, China.,Key Laboratory of Myopia, Ministry of HealthShanghai, China
| | - Wan-Jing Huang
- Eye Institute, Eye and ENT Hospital, College of Medicine, Fudan UniversityShanghai, China
| | - Min Wang
- Eye Institute, Eye and ENT Hospital, College of Medicine, Fudan UniversityShanghai, China
| | - Jun-Yi Chen
- Eye Institute, Eye and ENT Hospital, College of Medicine, Fudan UniversityShanghai, China
| | - Xing-Huai Sun
- Eye Institute, Eye and ENT Hospital, College of Medicine, Fudan UniversityShanghai, China.,State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan UniversityShanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality,Shanghai, China.,Key Laboratory of Myopia, Ministry of HealthShanghai, China
| | - Ji-Hong Wu
- Eye Institute, Eye and ENT Hospital, College of Medicine, Fudan UniversityShanghai, China.,State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan UniversityShanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality,Shanghai, China.,Key Laboratory of Myopia, Ministry of HealthShanghai, China
| |
Collapse
|
83
|
Nuzzi R, Tridico F. Glaucoma: Biological Trabecular and Neuroretinal Pathology with Perspectives of Therapy Innovation and Preventive Diagnosis. Front Neurosci 2017; 11:494. [PMID: 28928631 PMCID: PMC5591842 DOI: 10.3389/fnins.2017.00494] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 08/22/2017] [Indexed: 12/14/2022] Open
Abstract
Glaucoma is a common degenerative disease affecting retinal ganglion cells (RGC) and optic nerve axons, with progressive and chronic course. It is one of the most important reasons of social blindness in industrialized countries. Glaucoma can lead to the development of irreversible visual field loss, if not treated. Diagnosis may be difficult due to lack of symptoms in early stages of disease. In many cases, when patients arrive at clinical evaluation, a severe neuronal damage may have already occurred. In recent years, newer perspective in glaucoma treatment have emerged. The current research is focusing on finding newer drugs and associations or better delivery systems in order to improve the pharmacological treatment and patient compliance. Moreover, the application of various stem cell types with restorative and neuroprotective intent may be found appealing (intravitreal autologous cellular therapy). Advances are made also in terms of parasurgical treatment, characterized by various laser types and techniques. Moreover, recent research has led to the development of central and peripheral retinal rehabilitation (featuring residing cells reactivation and replacement of defective elements), as well as innovations in diagnosis through more specific and refined methods and inexpensive tests.
Collapse
Affiliation(s)
- Raffaele Nuzzi
- Eye Clinic Section, Department of Surgical Sciences, University of Turin, Ophthalmic HospitalTurin, Italy
| | - Federico Tridico
- Eye Clinic Section, Department of Surgical Sciences, University of Turin, Ophthalmic HospitalTurin, Italy
| |
Collapse
|
84
|
He S, Stankowska DL, Ellis DZ, Krishnamoorthy RR, Yorio T. Targets of Neuroprotection in Glaucoma. J Ocul Pharmacol Ther 2017; 34:85-106. [PMID: 28820649 DOI: 10.1089/jop.2017.0041] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Progressive neurodegeneration of the optic nerve and the loss of retinal ganglion cells is a hallmark of glaucoma, the leading cause of irreversible blindness worldwide, with primary open-angle glaucoma (POAG) being the most frequent form of glaucoma in the Western world. While some genetic mutations have been identified for some glaucomas, those associated with POAG are limited and for most POAG patients, the etiology is still unclear. Unfortunately, treatment of this neurodegenerative disease and other retinal degenerative diseases is lacking. For POAG, most of the treatments focus on reducing aqueous humor formation, enhancing uveoscleral or conventional outflow, or lowering intraocular pressure through surgical means. These efforts, in some cases, do not always lead to a prevention of vision loss and therefore other strategies are needed to reduce or reverse the progressive neurodegeneration. In this review, we will highlight some of the ocular pharmacological approaches that are being tested to reduce neurodegeneration and provide some form of neuroprotection.
Collapse
Affiliation(s)
- Shaoqing He
- North Texas Eye Research Institute, University of North Texas Health Science Center , Fort Worth, Texas
| | - Dorota L Stankowska
- North Texas Eye Research Institute, University of North Texas Health Science Center , Fort Worth, Texas
| | - Dorette Z Ellis
- North Texas Eye Research Institute, University of North Texas Health Science Center , Fort Worth, Texas
| | - Raghu R Krishnamoorthy
- North Texas Eye Research Institute, University of North Texas Health Science Center , Fort Worth, Texas
| | - Thomas Yorio
- North Texas Eye Research Institute, University of North Texas Health Science Center , Fort Worth, Texas
| |
Collapse
|
85
|
Morrison JC, Cepurna WO, Tehrani S, Choe TE, Jayaram H, Lozano DC, Fortune B, Johnson EC. A Period of Controlled Elevation of IOP (CEI) Produces the Specific Gene Expression Responses and Focal Injury Pattern of Experimental Rat Glaucoma. Invest Ophthalmol Vis Sci 2017; 57:6700-6711. [PMID: 27942722 PMCID: PMC5156512 DOI: 10.1167/iovs.16-20573] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose We determine if several hours of controlled elevation of IOP (CEI) will produce the optic nerve head (ONH) gene expression changes and optic nerve (ON) damage pattern associated with early experimental glaucoma in rats. Methods The anterior chambers of anesthetized rats were cannulated and connected to a reservoir to elevate IOP. Physiologic parameters were monitored. Following CEI at various recovery times, ON cross-sections were graded for axonal injury. Anterior ONHs were collected at 0 hours to 10 days following CEI and RNA extracted for quantitative PCR measurement of selected messages. The functional impact of CEI was assessed by electroretinography (ERG). Results During CEI, mean arterial pressure (99 ± 6 mm Hg) and other physiologic parameters remained stable. An 8-hour CEI at 60 mm Hg produced significant focal axonal degeneration 10 days after exposure, with superior lesions in 83% of ON. Message analysis in CEI ONH demonstrated expression responses previously identified in minimally injured ONH following chronic IOP elevation, as well as their sequential patterns. Anesthesia with cannulation at 20 mm Hg did not alter these message levels. Electroretinographic A- and B-waves, following a significant reduction at 2 days after CEI, were fully recovered at 2 weeks, while peak scotopic threshold response (pSTR) remained mildly but significantly depressed. Conclusions A single CEI reproduces ONH message changes and patterns of ON injury previously observed with chronic IOP elevation. Controlled elevation of IOP can allow detailed determination of ONH cellular and functional responses to an injurious IOP insult and provide a platform for developing future therapeutic interventions.
Collapse
Affiliation(s)
- John C Morrison
- The Kenneth C. Swan Ocular Neurobiology Laboratory, Casey Eye Institute, Oregon Health and Science University, Portland, Oregon, United States
| | - William O Cepurna
- The Kenneth C. Swan Ocular Neurobiology Laboratory, Casey Eye Institute, Oregon Health and Science University, Portland, Oregon, United States
| | - Shandiz Tehrani
- The Kenneth C. Swan Ocular Neurobiology Laboratory, Casey Eye Institute, Oregon Health and Science University, Portland, Oregon, United States
| | - Tiffany E Choe
- The Kenneth C. Swan Ocular Neurobiology Laboratory, Casey Eye Institute, Oregon Health and Science University, Portland, Oregon, United States
| | - Hari Jayaram
- The Kenneth C. Swan Ocular Neurobiology Laboratory, Casey Eye Institute, Oregon Health and Science University, Portland, Oregon, United States 2Glaucoma Service, NIHR Moorfields Biomedical Research Centre, London, United Kingdom
| | - Diana C Lozano
- The Kenneth C. Swan Ocular Neurobiology Laboratory, Casey Eye Institute, Oregon Health and Science University, Portland, Oregon, United States
| | - Brad Fortune
- Devers Eye Institute, Portland, Oregon, United States
| | - Elaine C Johnson
- The Kenneth C. Swan Ocular Neurobiology Laboratory, Casey Eye Institute, Oregon Health and Science University, Portland, Oregon, United States
| |
Collapse
|
86
|
Feng L, Chen H, Yi J, Troy JB, Zhang HF, Liu X. Long-Term Protection of Retinal Ganglion Cells and Visual Function by Brain-Derived Neurotrophic Factor in Mice With Ocular Hypertension. Invest Ophthalmol Vis Sci 2017; 57:3793-802. [PMID: 27421068 PMCID: PMC4961002 DOI: 10.1167/iovs.16-19825] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Purpose Glaucoma, frequently associated with elevated intraocular pressure (IOP), is characterized by progressive retinal ganglion cell (RGC) death and vision loss. Brain-derived neurotrophic factor (BDNF) has been studied as a candidate for neuroprotection in rodent models of experimental glaucoma, yet it remains to be determined whether BDNF exerts long-term protection for subtype RGCs and vision against chronic IOP elevation. Methods We induced modest and sustained IOP elevation by laser illumination and microbead injection in mice. Using a tamoxifen-induced Cre recombinase system, BDNF was upregulated in the mouse retina when sustained IOP elevation was induced. We then examined whether overexpression of BDNF protected RGCs and vision during the period of ocular hypertension. Given that BDNF modulates axon growth and dendritic formation in a subtype-dependent manner, we tested whether BDNF protects RGC dendritic structure against the hypertensive insult also in a subtype-dependent manner. Results Sustained IOP elevation was induced and lasted up to 6 months. Overexpression of BDNF delayed progressive RGC and axon loss in hypertensive eyes. Brain-derived neurotrophic factor overexpression also helped to preserve acuity against the chronic hypertensive insult. We classified RGCs into ON and ON–OFF subtypes based on their dendritic lamination pattern in the inner plexiform layer and found that BDNF prevented ON–RGC dendritic degeneration in mice with sustained ocular hypertension. Conclusions Our data demonstrated that BDNF can protect the dendritic fields of ON RGCs and reduce RGC and vision loss in mice with sustained ocular hypertension.
Collapse
Affiliation(s)
- Liang Feng
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States 2Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, Illinois, United States
| | - Hui Chen
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Ji Yi
- Department of Biomedical Engineering, Robert R. McCormick School of Engineering and Applied Science, Northwestern University, Evanston, Illinois, United States
| | - John B Troy
- Department of Biomedical Engineering, Robert R. McCormick School of Engineering and Applied Science, Northwestern University, Evanston, Illinois, United States
| | - Hao F Zhang
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States 3Department of Biomedical Engineering, Robert R. McCormick School of Engineering and Applied Science, Northwestern University, Evanston, Il
| | - Xiaorong Liu
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States 2Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, Illinois, United States
| |
Collapse
|
87
|
Bucher F, Walz JM, Bühler A, Aguilar E, Lange C, Diaz-Aguilar S, Martin G, Schlunck G, Agostini H, Friedlander M, Stahl A. CNTF Attenuates Vasoproliferative Changes Through Upregulation of SOCS3 in a Mouse-Model of Oxygen-Induced Retinopathy. Invest Ophthalmol Vis Sci 2017; 57:4017-26. [PMID: 27494343 PMCID: PMC4986766 DOI: 10.1167/iovs.15-18508] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Purpose Retinal vascular disease represents a major cause for vision loss in the Western world. Recent research has shown that neuronal and vascular damage are closely related in retinal disease. Ciliary neurotrophic factor (CNTF) is a well-studied neurotrophic factor that is currently being tested in clinical trials for the treatment of retinal degenerative diseases and macular telangiectasia. However, little is known about its effect on retinal vasculature. In this study, we investigate the effects of CNTF in retinal neovascular disease using the mouse model of oxygen-induced retinopathy (OIR). Methods Newborn pups were exposed to 75% oxygen from postnatal day (P)7 to P12 and subsequently returned to room air. Ciliary neurotrophic factor was injected intravitreally at OIR P12 and the vaso-obliterated and neovascular areas were quantified at OIR P17. Immunohistochemistry, RNA, and protein analysis were used to identify CNTF-responsive cells. In vitro experiments were performed to analyze the effect of CNTF on endothelial and astroglial cells. Results In the OIR model, CNTF facilitated capillary regrowth and attenuated preretinal neovascularization in a dose-dependent manner. The protective effect of CNTF was mediated via activation of the JAK/STAT3/SOCS3 signaling pathway. Immunohistochemical studies identified endothelial cells among others as CNTF-responsive cells in the retina. In vitro studies confirmed the anti-angiogenic effect of CNTF on endothelial cell sprouting. Conclusions This study provides evidence for a therapeutic potential of CNTF beyond degenerative retinal disease. Vasoproliferative retinopathies may benefit from a CNTF-dependent and SOCS3-mediated angiomodulatory effect.
Collapse
Affiliation(s)
- Felicitas Bucher
- Eye Center Medical Center, Faculty of Medicine, University of Freiburg, Germany 2Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California, United States
| | - Johanna M Walz
- Eye Center Medical Center, Faculty of Medicine, University of Freiburg, Germany 3Department of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany
| | - Anima Bühler
- Eye Center Medical Center, Faculty of Medicine, University of Freiburg, Germany
| | - Edith Aguilar
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California, United States
| | - Clemens Lange
- Eye Center Medical Center, Faculty of Medicine, University of Freiburg, Germany
| | - Sophia Diaz-Aguilar
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California, United States
| | - Gottfried Martin
- Eye Center Medical Center, Faculty of Medicine, University of Freiburg, Germany
| | - Günther Schlunck
- Eye Center Medical Center, Faculty of Medicine, University of Freiburg, Germany
| | - Hansjürgen Agostini
- Eye Center Medical Center, Faculty of Medicine, University of Freiburg, Germany
| | - Martin Friedlander
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California, United States
| | - Andreas Stahl
- Eye Center Medical Center, Faculty of Medicine, University of Freiburg, Germany
| |
Collapse
|
88
|
Overexpression of Brain-Derived Neurotrophic Factor Protects Large Retinal Ganglion Cells After Optic Nerve Crush in Mice. eNeuro 2017; 4:eN-NWR-0331-16. [PMID: 28101532 PMCID: PMC5240030 DOI: 10.1523/eneuro.0331-16.2016] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/21/2016] [Accepted: 12/28/2016] [Indexed: 12/18/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF), a neurotrophin essential for neuron survival and function, plays an important role in neuroprotection during neurodegenerative diseases. In this study, we examined whether a modest increase of retinal BDNF promotes retinal ganglion cell (RGC) survival after acute injury of the optic nerve in mice. We adopted an inducible Cre-recombinase transgenic system to up-regulate BDNF in the mouse retina and then examined RGC survival after optic nerve crush by in vivo imaging. We focused on one subtype of RGC with large soma expressing yellow fluorescent protein transgene that accounts for ∼11% of the total SMI-32-positive RGCs. The median survival time of this subgroup of SMI-32 cells was 1 week after nerve injury in control mice but 2 weeks when BDNF was up-regulated. Interestingly, we found that the survival time for RGCs taken as a whole was 2 weeks, suggesting that these large-soma RGCs are especially vulnerable to optic nerve crush injury. We also studied changes in axon number using confocal imaging, confirming first the progressive loss reported previously for wild-type mice and demonstrating that BDNF up-regulation extended axon survival. Together, our results demonstrate that the time course of RGC loss induced by optic nerve injury is type specific and that overexpression of BDNF prolongs the survival of one subgroup of SMI-32-positive RGCs.
Collapse
|
89
|
Protecting retinal ganglion cells. Eye (Lond) 2017; 31:218-224. [PMID: 28085136 DOI: 10.1038/eye.2016.299] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 11/26/2016] [Indexed: 01/08/2023] Open
Abstract
Retinal ganglion cell degeneration underlies several conditions which give rise to significant visual compromise, including glaucoma, hereditary optic neuropathies, ischaemic optic neuropathies, and demyelinating disease. In this review, we discuss the emerging strategies for neuroprotection specifically in the context of glaucoma, including pharmacological neuroprotection, mesenchymal stem cells, and gene therapy approaches. We highlight potential pitfalls that need to be considered when developing these strategies and outline future directions, including the prospects for clinical trials.
Collapse
|
90
|
Borrás T. The Pathway From Genes to Gene Therapy in Glaucoma: A Review of Possibilities for Using Genes as Glaucoma Drugs. Asia Pac J Ophthalmol (Phila) 2017; 6:80-93. [PMID: 28161916 PMCID: PMC6005701 DOI: 10.22608/apo.2016126] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 09/27/2016] [Indexed: 12/19/2022] Open
Abstract
Treatment of diseases with gene therapy is advancing rapidly. The use of gene therapy has expanded from the original concept of re-placing the mutated gene causing the disease to the use of genes to con-trol nonphysiological levels of expression or to modify pathways known to affect the disease. Genes offer numerous advantages over conventional drugs. They have longer duration of action and are more specific. Genes can be delivered to the target site by naked DNA, cells, nonviral, and viral vectors. The enormous progress of the past decade in molecular bi-ology and delivery systems has provided ways for targeting genes to the intended cell/tissue and safe, long-term vectors. The eye is an ideal organ for gene therapy. It is easily accessible and it is an immune-privileged site. Currently, there are clinical trials for diseases affecting practically every tissue of the eye, including those to restore vision in patients with Leber congenital amaurosis. However, the number of eye trials compared with those for systemic diseases is quite low (1.8%). Nevertheless, judg-ing by the vast amount of ongoing preclinical studies, it is expected that such number will increase considerably in the near future. One area of great need for eye gene therapy is glaucoma, where a long-term gene drug would eliminate daily applications and compliance issues. Here, we review the current state of gene therapy for glaucoma and the possibilities for treating the trabecular meshwork to lower intraocular pressure and the retinal ganglion cells to protect them from neurodegeneration.
Collapse
Affiliation(s)
- Teresa Borrás
- Department of Ophthalmology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
91
|
Krishnan A, Fei F, Jones A, Busto P, Marshak-Rothstein A, Ksander BR, Gregory-Ksander M. Overexpression of Soluble Fas Ligand following Adeno-Associated Virus Gene Therapy Prevents Retinal Ganglion Cell Death in Chronic and Acute Murine Models of Glaucoma. THE JOURNAL OF IMMUNOLOGY 2016; 197:4626-4638. [PMID: 27849168 DOI: 10.4049/jimmunol.1601488] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 10/15/2016] [Indexed: 12/28/2022]
Abstract
Glaucoma is a multifactorial disease resulting in the death of retinal ganglion cells (RGCs) and irreversible blindness. Glaucoma-associated RGC death depends on the proapoptotic and proinflammatory activity of membrane-bound Fas ligand (mFasL). In contrast to mFasL, the natural cleavage product, soluble Fas ligand (sFasL) inhibits mFasL-mediated apoptosis and inflammation and, therefore, is an mFasL antagonist. DBA/2J mice spontaneously develop glaucoma and, predictably, RGC destruction is exacerbated by expression of a mutated membrane-only FasL gene that lacks the extracellular cleavage site. Remarkably, one-time intraocular adeno-associated virus-mediated gene delivery of sFasL provides complete and sustained neuroprotection in the chronic DBA/2J and acute microbead-induced models of glaucoma, even in the presence of elevated intraocular pressure. This protection correlated with inhibition of glial activation, reduced production of TNF-α, and decreased apoptosis of RGCs and loss of axons. These data indicate that cleavage of FasL under homeostatic conditions, and the ensuing release of sFasL, normally limits the neurodestructive activity of FasL. The data further support the notion that sFasL, and not mFasL, contributes to the immune-privileged status of the eye.
Collapse
Affiliation(s)
- Anitha Krishnan
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114
| | - Fei Fei
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114.,Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, People's Republic of China; and
| | - Alexander Jones
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114
| | - Patricia Busto
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655
| | - Ann Marshak-Rothstein
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655
| | - Bruce R Ksander
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114
| | - Meredith Gregory-Ksander
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114;
| |
Collapse
|
92
|
Gupta V, Gupta VB, Chitranshi N, Gangoda S, Vander Wall R, Abbasi M, Golzan M, Dheer Y, Shah T, Avolio A, Chung R, Martins R, Graham S. One protein, multiple pathologies: multifaceted involvement of amyloid β in neurodegenerative disorders of the brain and retina. Cell Mol Life Sci 2016; 73:4279-4297. [PMID: 27333888 PMCID: PMC11108534 DOI: 10.1007/s00018-016-2295-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 06/14/2016] [Accepted: 06/15/2016] [Indexed: 01/18/2023]
Abstract
Accumulation of amyloid β (Aβ) and its aggregates in the ageing central nervous system is regarded synonymous to Alzheimer's disease (AD) pathology. Despite unquestionable advances in mechanistic and diagnostic aspects of the disease understanding, the primary cause of Aβ accumulation as well as its in vivo roles remains elusive; nonetheless, the majority of the efforts to address pathological mechanisms for therapeutic development are focused towards moderating Aβ accumulation in the brain. More recently, Aβ deposition has been identified in the eye and is linked with distinct age-related diseases including age-related macular degeneration, glaucoma as well as AD. Awareness of the Aβ accumulation in these markedly different degenerative disorders has led to an increasing body of work exploring overlapping mechanisms, a prospective biomarker role for Aβ and the potential to use retina as a model for brain related neurodegenerative disorders. Here, we present an integrated view of current understanding of the retinal Aβ deposition discussing the accumulation mechanisms, anticipated impacts and outlining ameliorative approaches that can be extrapolated to the retina for potential therapeutic benefits. Further longitudinal investigations in humans and animal models will determine retinal Aβ association as a potential pathognomonic, diagnostic or prognostic biomarker.
Collapse
Affiliation(s)
- Vivek Gupta
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Veer B Gupta
- School of Medical Sciences, Edith Cowan University, Perth, Australia.
| | - Nitin Chitranshi
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Sumudu Gangoda
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Roshana Vander Wall
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Mojdeh Abbasi
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Mojtaba Golzan
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Yogita Dheer
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Tejal Shah
- School of Medical Sciences, Edith Cowan University, Perth, Australia
| | - Alberto Avolio
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Roger Chung
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Ralph Martins
- School of Medical Sciences, Edith Cowan University, Perth, Australia
| | - Stuart Graham
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
- Save Sight Institute, Sydney University, Sydney, Australia
| |
Collapse
|
93
|
Weinreb RN, Leung CKS, Crowston JG, Medeiros FA, Friedman DS, Wiggs JL, Martin KR. Primary open-angle glaucoma. Nat Rev Dis Primers 2016; 2:16067. [PMID: 27654570 DOI: 10.1038/nrdp.2016.67] [Citation(s) in RCA: 286] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Glaucoma is an optic neuropathy that is characterized by the progressive degeneration of the optic nerve, leading to visual impairment. Glaucoma is the main cause of irreversible blindness worldwide, but typically remains asymptomatic until very severe. Open-angle glaucoma comprises the majority of cases in the United States and western Europe, of which, primary open-angle glaucoma (POAG) is the most common type. By contrast, in China and other Asian countries, angle-closure glaucoma is highly prevalent. These two types of glaucoma are characterized based on the anatomic configuration of the aqueous humour outflow pathway. The pathophysiology of POAG is not well understood, but it is an optic neuropathy that is thought to be associated with intraocular pressure (IOP)-related damage to the optic nerve head and resultant loss of retinal ganglion cells (RGCs). POAG is generally diagnosed during routine eye examination, which includes fundoscopic evaluation and visual field assessment (using perimetry). An increase in IOP, measured by tonometry, is not essential for diagnosis. Management of POAG includes topical drug therapies and surgery to reduce IOP, although new therapies targeting neuroprotection of RGCs and axonal regeneration are under development.
Collapse
Affiliation(s)
- Robert N Weinreb
- Shiley Eye Institute, Hamilton Glaucoma Center, Department of Ophthalmology, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, USA
| | - Christopher K S Leung
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jonathan G Crowston
- Department of Ophthalmology, Centre for Eye Research Australia, University of Melbourne, Melbourne, Victoria, Australia
| | - Felipe A Medeiros
- Shiley Eye Institute, Hamilton Glaucoma Center, Department of Ophthalmology, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, USA
| | - David S Friedman
- Dana Center for Preventive Ophthalmology, Johns Hopkins Wilmer Eye Institute, Baltimore, Maryland, USA
| | - Janey L Wiggs
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
| | - Keith R Martin
- Department of Ophthalmology and Cambridge NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK
| |
Collapse
|
94
|
Kimura A, Namekata K, Guo X, Harada C, Harada T. Neuroprotection, Growth Factors and BDNF-TrkB Signalling in Retinal Degeneration. Int J Mol Sci 2016; 17:ijms17091584. [PMID: 27657046 PMCID: PMC5037849 DOI: 10.3390/ijms17091584] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 09/01/2016] [Accepted: 09/14/2016] [Indexed: 12/18/2022] Open
Abstract
Neurotrophic factors play key roles in the development and survival of neurons. The potent neuroprotective effects of neurotrophic factors, including brain-derived neurotrophic factor (BDNF), ciliary neurotrophic factor (CNTF), glial cell-line derived neurotrophic factor (GDNF) and nerve growth factor (NGF), suggest that they are good therapeutic candidates for neurodegenerative diseases. Glaucoma is a neurodegenerative disease of the eye that causes irreversible blindness. It is characterized by damage to the optic nerve, usually due to high intraocular pressure (IOP), and progressive degeneration of retinal neurons called retinal ganglion cells (RGCs). Current therapy for glaucoma focuses on reduction of IOP, but neuroprotection may also be beneficial. BDNF is a powerful neuroprotective agent especially for RGCs. Exogenous application of BDNF to the retina and increased BDNF expression in retinal neurons using viral vector systems are both effective in protecting RGCs from damage. Furthermore, induction of BDNF expression by agents such as valproic acid has also been beneficial in promoting RGC survival. In this review, we discuss the therapeutic potential of neurotrophic factors in retinal diseases and focus on the differential roles of glial and neuronal TrkB in neuroprotection. We also discuss the role of neurotrophic factors in neuroregeneration.
Collapse
Affiliation(s)
- Atsuko Kimura
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.
| | - Kazuhiko Namekata
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.
| | - Xiaoli Guo
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.
| | - Chikako Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.
| | - Takayuki Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.
| |
Collapse
|
95
|
Wu N, Wang Y, Yang L, Cho KS. Signaling Networks of Retinal Ganglion Cell Formation and the Potential Application of Stem Cell–Based Therapy in Retinal Degenerative Diseases. Hum Gene Ther 2016; 27:609-20. [PMID: 27466076 DOI: 10.1089/hum.2016.083] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Affiliation(s)
- Nan Wu
- 1 Department of Ophthalmology, Southwest Eye Hospital, Southwest Hospital, Third Military Medical University , Chongqing, China
| | - Yi Wang
- 1 Department of Ophthalmology, Southwest Eye Hospital, Southwest Hospital, Third Military Medical University , Chongqing, China
| | - Lanbo Yang
- 2 Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Harvard Medical School , Boston, Massachusetts
| | - Kin-Sang Cho
- 2 Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Harvard Medical School , Boston, Massachusetts
| |
Collapse
|
96
|
Echevarria FD, Rickman AE, Sappington RM. Interleukin-6: A Constitutive Modulator of Glycoprotein 130, Neuroinflammatory and Cell Survival Signaling in Retina. ACTA ACUST UNITED AC 2016; 7. [PMID: 27747134 PMCID: PMC5061045 DOI: 10.4172/2155-9899.1000439] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Objective The interleukin-6 (IL-6) family of cytokines and their signal transducer glycoprotein (gp130) are implicated in inflammatory and cell survival functions in glaucoma. There are several avenues for interdependent modulation of IL-6 family members and gp130 signaling. Here we investigated whether IL-6 modulates gp130 and related neuroinflammatory, cell survival and regulatory signaling in both healthy and glaucomatous retina. Methods In naïve and glaucomatous (Microbead Occlusion Model), wildtype (WT) and IL-6 knockout (IL-6−/−) mice, we examined gp130 protein expression and localization, using western blot and immunohistochemistry. Gene targets related to IL-6 and gp130 signaling and pertinent to neuroinflammation (TNFα, IL-1β), cell health (Bax, Bcl-xl) and STAT3 regulation (Socs3) were quantified using qRTPCR. Results In the naïve retina, IL-6−/− retina contained significantly less gp130 compared to WT retina. This IL-6-related decrease in gp130 was accompanied by a reduction in mRNA expression of TNFα, Socs3 and Bax. After 4 weeks of microbead-induced ocular hypertension, both microbead- and saline-injected (control) eyes of IL-6−/− mice exhibited higher expression of TNFα, compared to WT mice. IL-1β expression was also reduced specifically in IL-6−/− retina with microbead-induced glaucoma. While saline and microbead injection increased Bcl-xl and Socs3 mRNA in both WT and IL-6−/− mice, IL-6−/− deficiency led to smaller increases for both Bcl-xl and Socs3. Conclusions Our findings support a role for IL-6 in setting baseline parameters for neuroinflammatory, cell health and gp130 regulatory signaling that can impact the nature and magnitude of retinal responses to glaucoma-related stressors.
Collapse
Affiliation(s)
| | - Abigayle E Rickman
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rebecca M Sappington
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Ophthalmology and Visual Sciences, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
97
|
Quigley HA. Understanding Glaucomatous Optic Neuropathy: The Synergy Between Clinical Observation and Investigation. Annu Rev Vis Sci 2016; 2:235-254. [PMID: 28532352 DOI: 10.1146/annurev-vision-111815-114417] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Glaucoma is a complex disorder of aging defined by the death of retinal ganglion cells and remodeling of connective tissues at the optic nerve head. Intraocular pressure-induced axonal injury at the optic nerve head leads to apoptosis. Loss of retinal ganglion cells follows a slowly progressive sequence. Clinical features of the disease have suggested and corroborated pathological events. The death of retinal ganglion cells causes secondary loss of neurons in the brain, but only as a by-product of injury to the retinal ganglion cells. Although therapy to lower intraocular pressure is moderately effective, new treatments are being developed to alter the remodeling of ocular connective tissue, to interrupt the injury signal from axon to soma, and to upregulate a variety of survival mechanisms.
Collapse
Affiliation(s)
- Harry A Quigley
- Glaucoma Center of Excellence, Wilmer Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287;
| |
Collapse
|
98
|
Glia-Neuron Interactions in the Retina Can Be Studied in Cocultures of Müller Cells and Retinal Ganglion Cells. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1087647. [PMID: 27429974 PMCID: PMC4939199 DOI: 10.1155/2016/1087647] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 04/27/2016] [Accepted: 05/19/2016] [Indexed: 11/18/2022]
Abstract
Glia-neuron partnership is important for inner retinal homeostasis and any disturbances may result in retinal ganglion cell (RGC) death. Müller cells support RGCs with essential functions such as removing excess glutamate and providing energy sources. The aim was to explore the impact of Müller cells on RGC survival. To investigate the Müller cell/RGC interactions we developed a coculture model, in which primary Müller cells were grown in inserts on top of pure primary RGC cultures. The impact of starvation and mitochondrial inhibition on the Müller cell ability to protect RGCs was studied. Moreover, the ability of Müller cells to remove glutamate from the extracellular space was investigated. RGC survival was evaluated by cell viability assays and glutamate uptake was assessed by kinetic uptake assays. We demonstrated a significantly increased RGC survival in presence of untreated and prestarved Müller cells. Additionally, prestarved Müller cells significantly increased RGC survival after mitochondrial inhibition. Finally, we revealed a significantly increased ability to take up glutamate in starved Müller cells. Overall, our study confirms essential roles of Müller cells in RGC survival. We suggest that targeting Müller cell function could have potential for future treatment strategies to prevent blinding neurodegenerative retinal diseases.
Collapse
|
99
|
Abstract
Over the last few years, huge progress has been made with regard to the understanding of molecular mechanisms underlying the pathogenesis of neurodegenerative diseases of the eye. Such knowledge has led to the development of gene therapy approaches to treat these devastating disorders. Challenges regarding the efficacy and efficiency of therapeutic gene delivery have driven the development of novel therapeutic approaches, which continue to evolve the field of ocular gene therapy. In this review article, we will discuss the evolution of preclinical and clinical strategies that have improved gene therapy in the eye, showing that treatment of vision loss has a bright future.
Collapse
Affiliation(s)
- Lolita Petit
- 1 Department of Ophthalmology and Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Hemant Khanna
- 1 Department of Ophthalmology and Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Department of Neurobiology, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Claudio Punzo
- 1 Department of Ophthalmology and Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Department of Neurobiology, University of Massachusetts Medical School , Worcester, Massachusetts
| |
Collapse
|
100
|
Retinal Macroglial Responses in Health and Disease. BIOMED RESEARCH INTERNATIONAL 2016; 2016:2954721. [PMID: 27294114 PMCID: PMC4887628 DOI: 10.1155/2016/2954721] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 04/14/2016] [Indexed: 12/20/2022]
Abstract
Due to their permanent and close proximity to neurons, glial cells perform essential tasks for the normal physiology of the retina. Astrocytes and Müller cells (retinal macroglia) provide physical support to neurons and supplement them with several metabolites and growth factors. Macroglia are involved in maintaining the homeostasis of extracellular ions and neurotransmitters, are essential for information processing in neural circuits, participate in retinal glucose metabolism and in removing metabolic waste products, regulate local blood flow, induce the blood-retinal barrier (BRB), play fundamental roles in local immune response, and protect neurons from oxidative damage. In response to polyetiological insults, glia cells react with a process called reactive gliosis, seeking to maintain retinal homeostasis. When malfunctioning, macroglial cells can become primary pathogenic elements. A reactive gliosis has been described in different retinal pathologies, including age-related macular degeneration (AMD), diabetes, glaucoma, retinal detachment, or retinitis pigmentosa. A better understanding of the dual, neuroprotective, or cytotoxic effect of macroglial involvement in retinal pathologies would help in treating the physiopathology of these diseases. The extensive participation of the macroglia in retinal diseases points to these cells as innovative targets for new drug therapies.
Collapse
|