1
|
Servillo A, Sacconi R, Oldoni G, Barlocci E, Tombolini B, Battista M, Fantaguzzi F, Rissotto F, Mularoni C, Parravano M, Zucchiatti I, Querques L, Bandello F, Querques G. Advancements in Imaging and Therapeutic Options for Dry Age-Related Macular Degeneration and Geographic Atrophy. Ophthalmol Ther 2024; 13:2067-2082. [PMID: 38833127 PMCID: PMC11246354 DOI: 10.1007/s40123-024-00970-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/13/2024] [Indexed: 06/06/2024] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of vision loss in the elderly, with dry AMD (d-AMD) leading to geographic atrophy (GA) and significant visual impairment. Multimodal imaging plays a crucial role in d-AMD diagnosis and management, allowing for detailed classification of patient phenotypes and aiding in treatment planning and prognosis determination. Treatment approaches for d-AMD have recently witnessed profound change with the development of specific drugs targeting the complement cascade, with the first anticomplement agents recently approved for GA treatment. Additionally, emerging strategies such as gene therapy and laser treatments may offer potential benefits, though further research is needed to fully establish their efficacy. However, the lack of effective therapies capable of restoring damaged retinal cells remains a major challenge. In the future, genetic treatments aimed at preventing the progression of d-AMD may emerge as a powerful approach. Currently, however, their development is still in the early stages.
Collapse
Affiliation(s)
- Andrea Servillo
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Riccardo Sacconi
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Gloria Oldoni
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Eugenio Barlocci
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Beatrice Tombolini
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Marco Battista
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Federico Fantaguzzi
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Federico Rissotto
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Cecilia Mularoni
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | | | - Ilaria Zucchiatti
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Lea Querques
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Francesco Bandello
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Giuseppe Querques
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy.
- Division of Head and Neck, Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy.
| |
Collapse
|
2
|
Miralles de Imperial-Ollero JA, Gallego-Ortega A, Ortín-Martínez A, Villegas-Pérez MP, Valiente-Soriano FJ, Vidal-Sanz M. Animal Models of LED-Induced Phototoxicity. Short- and Long-Term In Vivo and Ex Vivo Retinal Alterations. Life (Basel) 2021; 11:life11111137. [PMID: 34833013 PMCID: PMC8617611 DOI: 10.3390/life11111137] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 12/26/2022] Open
Abstract
Phototoxicity animal models have been largely studied due to their degenerative communalities with human pathologies, e.g., age-related macular degeneration (AMD). Studies have documented not only the effects of white light exposure, but also other wavelengths using LEDs, such as blue or green light. Recently, a blue LED-induced phototoxicity (LIP) model has been developed that causes focal damage in the outer layers of the superior-temporal region of the retina in rodents. In vivo studies described a progressive reduction in retinal thickness that affected the most extensively the photoreceptor layer. Functionally, a transient reduction in a- and b-wave amplitude of the ERG response was observed. Ex vivo studies showed a progressive reduction of cones and an involvement of retinal pigment epithelium cells in the area of the lesion and, in parallel, an activation of microglial cells that perfectly circumscribe the damage in the outer retinal layer. The use of neuroprotective strategies such as intravitreal administration of trophic factors, e.g., basic fibroblast growth factor (bFGF), brain-derived neurotrophic factor (BDNF), ciliary neurotrophic factor (CNTF) or pigment epithelium-derived factor (PEDF) and topical administration of the selective alpha-2 agonist (Brimonidine) have demonstrated to increase the survival of the cone population after LIP.
Collapse
Affiliation(s)
- Juan A. Miralles de Imperial-Ollero
- Departamento de Oftalmología, Universidad de Murcia e Instituto Murciano de Investigación Biosanitaria (IMIB) Virgen de la Arrixaca, Campus de CC de la Salud, El Palmar, 30120 Murcia, Spain; (J.A.M.d.I.-O.); (A.G.-O.); (M.P.V.-P.)
| | - Alejandro Gallego-Ortega
- Departamento de Oftalmología, Universidad de Murcia e Instituto Murciano de Investigación Biosanitaria (IMIB) Virgen de la Arrixaca, Campus de CC de la Salud, El Palmar, 30120 Murcia, Spain; (J.A.M.d.I.-O.); (A.G.-O.); (M.P.V.-P.)
| | - Arturo Ortín-Martínez
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON M5T 2S8, Canada;
| | - María Paz Villegas-Pérez
- Departamento de Oftalmología, Universidad de Murcia e Instituto Murciano de Investigación Biosanitaria (IMIB) Virgen de la Arrixaca, Campus de CC de la Salud, El Palmar, 30120 Murcia, Spain; (J.A.M.d.I.-O.); (A.G.-O.); (M.P.V.-P.)
| | - Francisco J. Valiente-Soriano
- Departamento de Oftalmología, Universidad de Murcia e Instituto Murciano de Investigación Biosanitaria (IMIB) Virgen de la Arrixaca, Campus de CC de la Salud, El Palmar, 30120 Murcia, Spain; (J.A.M.d.I.-O.); (A.G.-O.); (M.P.V.-P.)
- Correspondence: (F.J.V.-S.); (M.V.-S.); Tel.: +34-868-88-4503 (F.J.V-S.); +34-868-88-4330 (M.V.-S.)
| | - Manuel Vidal-Sanz
- Departamento de Oftalmología, Universidad de Murcia e Instituto Murciano de Investigación Biosanitaria (IMIB) Virgen de la Arrixaca, Campus de CC de la Salud, El Palmar, 30120 Murcia, Spain; (J.A.M.d.I.-O.); (A.G.-O.); (M.P.V.-P.)
- Correspondence: (F.J.V.-S.); (M.V.-S.); Tel.: +34-868-88-4503 (F.J.V-S.); +34-868-88-4330 (M.V.-S.)
| |
Collapse
|
3
|
Ghasemi M, Alizadeh E, Motlagh BF, Zarghami N. The effect of exogenous ciliary neurotrophic factor on cell cycle and neural differentiation markers of in vitro model cells: New insights for future therapeutic approaches. Cell Biochem Funct 2021; 39:636-645. [PMID: 33890305 DOI: 10.1002/cbf.3628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/12/2021] [Accepted: 02/16/2021] [Indexed: 11/06/2022]
Abstract
Retinoblastoma is known as childhood rare malignancy of the retina. Ciliary neurotrophic factor (CNTF) was previously found to reduce degeneration and promote retina survival. This work investigated the effects of CNTF supplementation on in-vitro model cells including retinoblastoma (Y79) and adipose-derived mesenchymal stem cells (AMSCs) viability, proliferation, gene expression and cell cycle. A drop of viability was detected in Y79 treated with CNTF in a dose-dependent manner (P < .05). However, the proliferation of AMSCs was increased at lower concentrations of CNTF (5 ng/mL), but declined in higher doses (50 and 100 ng/mL). The BrdU assay confirmed the MTT assay results. Cell cycle was arrested in both Y79 and AMSCs in the G0/G1 phase by CNTF treatment. A considerable down-regulation of Bcl2, CycD1 and N-Myc genes expression (P < .05) inversely, P15 and P21 genes up-regulation in treated Y79 cells was observed. Besides, stemness genes' transcription was reduced in AMSCs (P < .05), and levels of neuronal-specific markers such as neuron-specific enolase (NSE) and neuronal nuclei (NeuN) were increased (P < .05). The findings of this study suggest a promising potential of CNTF in terms of arresting Y79 retinoblastoma cells, and differentiation-inducing to AMSCs, which could be valuable for managing future innovative treatments targeting retinoblastoma. SIGNIFICANCE OF THE STUDY: We demonstrate that CNTF has the potential to reduce proliferation of Y79 cells and induce the cell cycle arrest of them. Also, down-regulation of oncogenes (such as N-Myc) while up-regulation of tumour suppressor genes (such as P21) was detected by exposure of Y79 cells to CNTF. Furthermore, we observed the cell cycle arrest, reduction of stemness gene and up-regulation of neural differentiation markers in AMSCs treated with CNTF. These results support the probable promising effects of CNTF for controlling retinoblastoma.
Collapse
Affiliation(s)
- Maryam Ghasemi
- Stem Cell Research Center (SRC), Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Effat Alizadeh
- Stem Cell Research Center (SRC), Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Fallahi Motlagh
- Department of Ophthalmology, Nikokar Eye Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nosratollah Zarghami
- Stem Cell Research Center (SRC), Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
4
|
Potential neuroprotective biomolecules in ophthalmology. Int Ophthalmol 2020; 41:1103-1109. [PMID: 33180279 DOI: 10.1007/s10792-020-01634-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 10/29/2020] [Indexed: 10/23/2022]
Abstract
PURPOSES Retinal neurodegenerative diseases are responsible for a huge number of ocular problems worldwide. It seems that the progression of these diseases can be managed by the application of neuroprotective molecules particularly in the early stages. This article focuses on the most common neuroprotective bioagents under investigation in ophthalmology. METHODS We searched the web of science, PubMed and Scopus databases with these keywords: "glaucoma," "diabetic retinopathy," "age-related macular degeneration," "optic neuropathy and retinal degeneration" and/or "neuroprotection." RESULTS The most commonly utilized neuroprotective drugs for ophthalmology diseases were introduced in this study. It seems that these agents can be divided into three categories according to their mechanism of action: (A) neurotrophins, (B) decreasing effect on intraocular pressure and (C) inhibition of retinal neuron apoptosis. CONCLUSION A broad range of drugs has been illustrated in the literature for treatment of neuro-ophthalmic diseases. A good classification of the most applied drugs in this field can help specialists to prescribe the best matched drug considering the stage and progression of disease. However, controlled clinical trials are needed for better evaluation of the effects of these products.
Collapse
|
5
|
Ao J, Chidlow G, Wood JPM, Casson RJ. Safety Profile of Slit-Lamp-Delivered Retinal Laser Photobiomodulation. Transl Vis Sci Technol 2020; 9:22. [PMID: 32818109 PMCID: PMC7396177 DOI: 10.1167/tvst.9.4.22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 01/10/2020] [Indexed: 01/15/2023] Open
Abstract
Purpose Photobiomodulation (PBM) refers to therapeutic irradiation of tissue with low-energy, 630- to 1000-nm wavelength light. An increasing body of evidence supports a beneficial effect of PBM in retinal disorders. To date, most studies have utilized light-emitting diode irradiation sources. Slit-lamp-mounted retinal lasers produce a coherent beam that can be delivered with precisely defined dosages and predetermined target area; however, the use of retinal lasers raises safety concerns that warrant investigation prior to clinical application. In this study, we determined safe dosages of laser-delivered PBM to the retina. Methods A custom-designed, slit-lamp-delivered, 670-nm, red/near-infrared laser was used to administer a range of irradiances to healthy pigmented and non-pigmented rat retinas. The effects of PBM on various functional and structural parameters of the retina were evaluated utilizing a combination of electroretinography, Spectral Domain Optical Coherence (SD-OCT), fluorescein angiography, histology and immunohistochemistry. Results In non-pigmented rats, no adverse events were identified at any irradiances up to 500 mW/cm2. In pigmented rats, no adverse events were identified at irradiances of 25 or 100 mW/cm2; however, approximately one-third of rats that received 500 mW/cm2 displayed very localized photoreceptor damage in the peripapillary region, typically adjacent to the optic nerve head. Conclusions A safety threshold exists for laser-delivered PBM in pigmented retinas and was identified as 500 mW/cm2 irradiance; therefore, caution should be exercised in the dosage of laser-delivered PBM administered to pigmented retinas. Translational Relevance This study provides important data necessary for clinical translation of laser-delivered PBM for retinal diseases.
Collapse
Affiliation(s)
- Jack Ao
- Ophthalmic Research Laboratories, Discipline of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Glyn Chidlow
- Ophthalmic Research Laboratories, Discipline of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - John P M Wood
- Ophthalmic Research Laboratories, Discipline of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Robert J Casson
- Ophthalmic Research Laboratories, Discipline of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
6
|
Li S, Sato K, Gordon WC, Sendtner M, Bazan NG, Jin M. Ciliary neurotrophic factor (CNTF) protects retinal cone and rod photoreceptors by suppressing excessive formation of the visual pigments. J Biol Chem 2018; 293:15256-15268. [PMID: 30115683 DOI: 10.1074/jbc.ra118.004008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 08/15/2018] [Indexed: 12/25/2022] Open
Abstract
The retinal pigment epithelium (RPE)-dependent visual cycle provides 11-cis-retinal to opsins in the photoreceptor outer segments to generate functional visual pigments that initiate phototransduction in response to light stimuli. Both RPE65 isomerase of the visual cycle and the rhodopsin visual pigment have recently been identified as critical players in mediating light-induced retinal degeneration. These findings suggest that the expression and function of RPE65 and rhodopsin need to be coordinately controlled to sustain normal vision and to protect the retina from photodamage. However, the mechanism controlling the development of the retinal visual system remains poorly understood. Here, we show that deficiency in ciliary neurotrophic factor (CNTF) up-regulates the levels of rod and cone opsins accompanied by an increase in the thickness of the outer nuclear layers and the lengths of cone and rod outer segments in the mouse retina. Moreover, retinoid isomerase activity, expression levels of RPE65 and lecithin:retinol acyltransferase (LRAT), which synthesizes the RPE65 substrate, were also significantly increased in the Cntf -/- RPE. Rod a-wave and cone b-wave amplitudes of electroretinograms were increased in Cntf -/- mice, but rod b-wave amplitudes were unchanged compared with those in WT mice. Up-regulated RPE65 and LRAT levels accelerated both the visual cycle rate and recovery rate of rod light sensitivity in Cntf -/- mice. Of note, rods and cones in Cntf -/- mice exhibited hypersusceptibility to light-induced degeneration. These results indicate that CNTF is a common extracellular factor that prevents excessive production of opsins, the photoreceptor outer segments, and 11-cis-retinal to protect rods and cones from photodamage.
Collapse
Affiliation(s)
- Songhua Li
- From the Neuroscience Center of Excellence and
| | - Kota Sato
- From the Neuroscience Center of Excellence and
| | - William C Gordon
- From the Neuroscience Center of Excellence and.,Department of Ophthalmology, Louisiana State University School of Medicine, New Orleans, Louisiana 70112 and
| | - Michael Sendtner
- the Institute of Clinical Neurobiology, University Hospital Würzburg, D-97078 Würzburg, Germany
| | - Nicolas G Bazan
- From the Neuroscience Center of Excellence and.,Department of Ophthalmology, Louisiana State University School of Medicine, New Orleans, Louisiana 70112 and
| | - Minghao Jin
- From the Neuroscience Center of Excellence and .,Department of Ophthalmology, Louisiana State University School of Medicine, New Orleans, Louisiana 70112 and
| |
Collapse
|
7
|
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: 1.1] [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
|
8
|
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: 1.9] [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
|
9
|
Espino Barros A, Amram AL, Derham AM, Smith SV, Lee AG. Management of ischemic optic neuropathies. EXPERT REVIEW OF OPHTHALMOLOGY 2017. [DOI: 10.1080/17469899.2017.1291341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Angelina Espino Barros
- Department of Ophthalmology, Centro Médico Zambrano Hellion, San Pedro Garza García, México
| | - Alec L. Amram
- Department of Ophthalmology, University of Texas Medical Branch, Galveston, TX, USA
| | - Angeline Mariani Derham
- Department of Ophthalmology, University of Texas Health Science Center San Antonio School of Medicine, San Antonio, TX, USA
| | - Stacy V. Smith
- Department of Ophthalmology, Blanton Eye Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Andrew G. Lee
- Department of Ophthalmology, University of Texas Medical Branch, Galveston, TX, USA
- Department of Ophthalmology, Blanton Eye Institute, Houston Methodist Hospital, Houston, TX, USA
- Departments of Ophthalmology, Neurology, and Neurosurgery, Weill Cornell Medicine, New York City, NY, USA
- Section of Ophthalmology, UT MD Anderson Cancer Center, Houston, TX, USA
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
10
|
Becker S, Eastlake K, Jayaram H, Jones MF, Brown RA, McLellan GJ, Charteris DG, Khaw PT, Limb GA. Allogeneic Transplantation of Müller-Derived Retinal Ganglion Cells Improves Retinal Function in a Feline Model of Ganglion Cell Depletion. Stem Cells Transl Med 2016; 5:192-205. [PMID: 26718648 PMCID: PMC4729554 DOI: 10.5966/sctm.2015-0125] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 11/11/2015] [Indexed: 11/16/2022] Open
Abstract
Human Müller glia with stem cell characteristics (hMGSCs) have been shown to improve retinal function upon transplantation into rat models of retinal ganglion cell (RGC) depletion. However, their translational potential may depend upon successful engraftment and improvement of retinal function in experimental models with anatomical and functional features resembling those of the human eye. We investigated the effect of allogeneic transplantation of feline Müller glia with the ability to differentiate into cells expressing RGC markers, following ablation of RGCs by N-methyl-d-aspartate (NMDA). Unlike previous observations in the rat, transplantation of hMGSC-derived RGCs into the feline vitreous formed aggregates and elicited a severe inflammatory response without improving visual function. In contrast, allogeneic transplantation of feline MGSC (fMGSC)-derived RGCs into the vitrectomized eye improved the scotopic threshold response (STR) of the electroretinogram (ERG). Despite causing functional improvement, the cells did not attach onto the retina and formed aggregates on peripheral vitreous remnants, suggesting that vitreous may constitute a barrier for cell attachment onto the retina. This was confirmed by observations that cellular scaffolds of compressed collagen and enriched preparations of fMGSC-derived RGCs facilitated cell attachment. Although cells did not migrate into the RGC layer or the optic nerve, they significantly improved the STR and the photopic negative response of the ERG, indicative of increased RGC function. These results suggest that MGSCs have a neuroprotective ability that promotes partial recovery of impaired RGC function and indicate that cell attachment onto the retina may be necessary for transplanted cells to confer neuroprotection to the retina. Significance: Müller glia with stem cell characteristics are present in the adult human retina, but they do not have regenerative ability. These cells, however, have potential for development of cell therapies to treat retinal disease. Using a feline model of retinal ganglion cell (RGC) depletion, cell grafting methods to improve RGC function have been developed. Using cellular scaffolds, allogeneic transplantation of Müller glia-derived RGC promoted cell attachment onto the retina and enhanced retinal function, as judged by improvement of the photopic negative and scotopic threshold responses of the electroretinogram. The results suggest that the improvement of RGC function observed may be ascribed to the neuroprotective ability of these cells and indicate that attachment of the transplanted cells onto the retina is required to promote effective neuroprotection.
Collapse
Affiliation(s)
- Silke Becker
- Institute of Ophthalmology, University College London, London, United Kingdom
| | - Karen Eastlake
- Institute of Ophthalmology, University College London, London, United Kingdom
| | - Hari Jayaram
- National Institute for Health Research Biomedical Research Centre for Ophthalmology, Institute of Ophthalmology, University College London, London, United Kingdom
| | - Megan F Jones
- Institute of Ophthalmology, University College London, London, United Kingdom
| | - Robert A Brown
- Institute of Orthopaedics and Musculoskeletal Science, University College London, London, United Kingdom
| | - Gillian J McLellan
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - David G Charteris
- National Institute for Health Research Biomedical Research Centre for Ophthalmology, Institute of Ophthalmology, University College London, London, United Kingdom
| | - Peng T Khaw
- National Institute for Health Research Biomedical Research Centre for Ophthalmology, Institute of Ophthalmology, University College London, London, United Kingdom
| | - G Astrid Limb
- National Institute for Health Research Biomedical Research Centre for Ophthalmology, Institute of Ophthalmology, University College London, London, United Kingdom
| |
Collapse
|
11
|
Agca C, Boldt K, Gubler A, Meneau I, Corpet A, Samardzija M, Stucki M, Ueffing M, Grimm C. Expression of leukemia inhibitory factor in Müller glia cells is regulated by a redox-dependent mRNA stability mechanism. BMC Biol 2015; 13:30. [PMID: 25907681 PMCID: PMC4462110 DOI: 10.1186/s12915-015-0137-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 04/08/2015] [Indexed: 12/15/2022] Open
Abstract
Background Photoreceptor degeneration is a main hallmark of many blinding diseases making protection of photoreceptors crucial to prevent vision loss. Thus, regulation of endogenous neuroprotective factors may be key for cell survival and attenuation of disease progression. Important neuroprotective factors in the retina include H2O2 generated by injured photoreceptors, and leukemia inhibitory factor (LIF) expressed in Müller glia cells in response to photoreceptor damage. Results We present evidence that H2O2 connects to the LIF response by inducing stabilization of Lif transcripts in Müller cells. This process was independent of active gene transcription and p38 MAPK, but relied on AU-rich elements (AREs), which we identified within the highly conserved Lif 3′UTR. Affinity purification combined with quantitative mass spectrometry identified several proteins that bound to these AREs. Among those, interleukin enhancer binding factor 3 (ILF3) was confirmed to participate in the redox-dependent Lif mRNA stabilization. Additionally we show that KH-type splicing regulatory protein (KHSRP) was crucial for maintaining basal Lif expression levels in non-stressed Müller cells. Conclusions Our results suggest that H2O2-induced redox signaling increases Lif transcript levels through ILF3 mediated mRNA stabilization. Generation of H2O2 by injured photoreceptors may thus enhance stability of Lif mRNA and therefore augment neuroprotective LIF signaling during degenerative conditions in vivo. Electronic supplementary material The online version of this article (doi:10.1186/s12915-015-0137-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Cavit Agca
- Department of Ophthalmology, Lab for Retinal Cell Biology, University of Zurich, Wagistrasse 14, Zurich, 8091, Switzerland. .,Present address: Department of Biomedicine, University Hospital Basel, Basel, 4031, Switzerland.
| | - Karsten Boldt
- Division of Experimental Ophthalmology and Medical Proteome Center, Centre for Ophthalmology, University of Tübingen, 72076, Tübingen, Germany.
| | - Andrea Gubler
- Department of Ophthalmology, Lab for Retinal Cell Biology, University of Zurich, Wagistrasse 14, Zurich, 8091, Switzerland.
| | - Isabelle Meneau
- Department of Ophthalmology, Lab for Retinal Cell Biology, University of Zurich, Wagistrasse 14, Zurich, 8091, Switzerland.
| | - Armelle Corpet
- Department of Gynecology, University of Zurich, Zurich, 8091, Switzerland. .,Present address: Center for Molecular and Cellular Physiology and Genetics, University Lyon I, Villeurbanne, France.
| | - Marijana Samardzija
- Department of Ophthalmology, Lab for Retinal Cell Biology, University of Zurich, Wagistrasse 14, Zurich, 8091, Switzerland.
| | - Manuel Stucki
- Department of Gynecology, University of Zurich, Zurich, 8091, Switzerland.
| | - Marius Ueffing
- Division of Experimental Ophthalmology and Medical Proteome Center, Centre for Ophthalmology, University of Tübingen, 72076, Tübingen, Germany.
| | - Christian Grimm
- Department of Ophthalmology, Lab for Retinal Cell Biology, University of Zurich, Wagistrasse 14, Zurich, 8091, Switzerland. .,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, 8091, Switzerland. .,Neuroscience Center (ZNZ), University of Zurich, Zurich, 8091, Switzerland.
| |
Collapse
|
12
|
|
13
|
Mathews MK, Guo Y, Langenberg P, Bernstein SL. Ciliary neurotrophic factor (CNTF)-mediated ganglion cell survival in a rodent model of non-arteritic anterior ischaemic optic neuropathy (NAION). Br J Ophthalmol 2014; 99:133-7. [PMID: 25336580 DOI: 10.1136/bjophthalmol-2014-305969] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
BACKGROUND Ciliary neurotrophic factor (CNTF) has been shown to protect retinal ganglion cells (RGCs) in traumatic optic nerve injury. We sought to evaluate this neuroprotective effect of CNTF after an ischaemic event using rodent anterior ischaemic optic neuropathy (rAION), a mouse model of non-arteritic anterior ischaemic optic neuropathy (NAION). METHODS We induced rAION in Thy1-cyan fluorescent protein (CFP) transgenic mice by exposing the optic nerve to frequency doubled neodymium yttrium aluminium garnet laser pulses following intravenous rose bengal injection. One day after rAION induction, an intravitreal injection of 0.75 μg CNTF or vehicle (sham injection) was given. Animals were euthanised on day 15 after induction, tissues isolated and CFP cells in the RGC layer were counted using stereology in flat-mounted retina. The average number of CFP-positive (CFP+) cells was determined for each study group and the percentages of RGC loss were compared between the different groups. RESULTS Two weeks after rAION induction, significantly more (CFP+) cells were preserved in CNTF-treated eyes than in sham-injected controls. Sham-treated animals showed a 58% loss of CFP+ cells. In contrast, CFP+ cell density in CNTF-treated eyes decreased by only 10%, when compared with untreated control eyes. This increased survival was statistically significant (p<0.05). CONCLUSIONS CNTF exerts a neuroprotective effect in ischaemic optic nerve injury and promotes RGC survival, suggesting that CNTF may be effective in the clinical treatment of human NAION.
Collapse
Affiliation(s)
- Michaela K Mathews
- Department of Ophthalmology and Visual Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yan Guo
- Department of Ophthalmology and Visual Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Patricia Langenberg
- Department of Epidemiology and Public Health, University of Maryland, Baltimore, Maryland, USA
| | - Steven L Bernstein
- Department of Ophthalmology and Visual Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
14
|
Dysregulation of neurotrophic and inflammatory systems accompanied by decreased CREB signaling in ischemic rat retina. Exp Eye Res 2014; 125:156-63. [PMID: 24954538 DOI: 10.1016/j.exer.2014.06.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 05/30/2014] [Accepted: 06/03/2014] [Indexed: 12/22/2022]
Abstract
Although permanent bilateral common carotid artery occlusion (2VO) has been demonstrated to induce retinal injury, there is still a lack of systematic research on the complex processing of retinal degeneration. In the present study, time-dependent (at three, 14, 60 days after 2VO surgery) changes of neurotrophic and inflammatory systems, as well as cAMP-responsive element binding protein (CREB) signaling, which has been previously reported to effectively regulate these two systems, were evaluated. First, a morphological study confirmed that 2VO surgery progressively induced severe inner retinal degeneration and down-regulation of synaptic proteins, PSD95 and synaptophysin. The mRNA or protein levels of neurotrophic factors (NGF, BDNF, NT-3 and GDNF) and their receptors (TrkA, TrkB and TrkC) showed marked and persistent down-regulation in the rat retina since three days after 2VO surgery, whereas the gene transcription levels of CNTF were increased and p75(NTR) mRNA levels remained unchanged. In contrast to inner retinal degeneration, retinal Müller cells displayed rapid and prolonged activation since three days after 2VO lesion, whereas the microglia cell number, and TNF-α and IL-1β levels showed a robust increase with a maximal effect at three days and returned to levels that were slightly over baseline at 14 and 60 days after 2VO lesion. Interestingly, the gene expression levels of iNOS significantly decreased in the rat retina at both three and 14 days after 2VO surgery. Finally, as we hypothesized, remarkable reduction of CREB and extracellular signal-regulated kinase (ERK) phosphorylation levels were observed in the rat retina at three days after 2VO surgery. Thus, for the first time, our study demonstrated that chronic ischemia induced long-term aberrant CREB signaling and time-dependent progressive dysregulation of neurotrophic and inflammatory systems in the retina, which may provide important clues for a better understanding of the pathogenesis of retinal ischemic damage.
Collapse
|
15
|
Agca C, Gubler A, Traber G, Beck C, Imsand C, Ail D, Caprara C, Grimm C. p38 MAPK signaling acts upstream of LIF-dependent neuroprotection during photoreceptor degeneration. Cell Death Dis 2013; 4:e785. [PMID: 24008729 PMCID: PMC3789181 DOI: 10.1038/cddis.2013.323] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 07/09/2013] [Accepted: 07/15/2013] [Indexed: 12/31/2022]
Abstract
In many blinding diseases of the retina, loss of function and thus severe visual impairment results from apoptotic cell death of damaged photoreceptors. In an attempt to survive, injured photoreceptors generate survival signals to induce intercellular protective mechanisms that eventually may rescue photoreceptors from entering an apoptotic death pathway. One such endogenous survival pathway is controlled by leukemia inhibitory factor (LIF), which is produced by a subset of Muller glia cells in response to photoreceptor injury. In the absence of LIF, survival components are not activated and photoreceptor degeneration is accelerated. Although LIF is a crucial factor for photoreceptor survival, the detailed mechanism of its induction in the retina has not been elucidated. Here, we show that administration of tumor necrosis factor-alpha (TNF) was sufficient to fully upregulate Lif expression in Muller cells in vitro and the retina in vivo. Increased Lif expression depended on p38 mitogen-activated protein kinase (MAPK) since inhibition of its activity abolished Lif expression in vitro and in vivo. Inhibition of p38 MAPK activity reduced the Lif expression also in the model of light-induced retinal degeneration and resulted in increased cell death in the light-exposed retina. Thus, expression of Lif in the injured retina and activation of the endogenous survival pathway involve signaling through p38 MAPK.
Collapse
Affiliation(s)
- C Agca
- Lab for Retinal Cell Biology, Department of Ophthalmology, University of Zurich, Zurich 8091, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Munemasa Y, Kitaoka Y. Molecular mechanisms of retinal ganglion cell degeneration in glaucoma and future prospects for cell body and axonal protection. Front Cell Neurosci 2013; 6:60. [PMID: 23316132 PMCID: PMC3540394 DOI: 10.3389/fncel.2012.00060] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Accepted: 12/06/2012] [Indexed: 12/20/2022] Open
Abstract
Glaucoma, which affects more than 70 million people worldwide, is a heterogeneous group of disorders with a resultant common denominator; optic neuropathy, eventually leading to irreversible blindness. The clinical manifestations of primary open-angle glaucoma (POAG), the most common subtype of glaucoma, include excavation of the optic disc and progressive loss of visual field. Axonal degeneration of retinal ganglion cells (RGCs) and apoptotic death of their cell bodies are observed in glaucoma, in which the reduction of intraocular pressure (IOP) is known to slow progression of the disease. A pattern of localized retinal nerve fiber layer (RNFL) defects in glaucoma patients indicates that axonal degeneration may precede RGC body death in this condition. The mechanisms of degeneration of neuronal cell bodies and their axons may differ. In this review, we addressed the molecular mechanisms of cell body death and axonal degeneration in glaucoma and proposed axonal protection in addition to cell body protection. The concept of axonal protection may become a new therapeutic strategy to prevent further axonal degeneration or revive dying axons in patients with preperimetric glaucoma. Further study will be needed to clarify whether the combination therapy of axonal protection and cell body protection will have greater protective effects in early or progressive glaucomatous optic neuropathy (GON).
Collapse
Affiliation(s)
- Yasunari Munemasa
- Department of Ophthalmology, St. Marianna University School of Medicine Kawasaki, Kanagawa, Japan
| | | |
Collapse
|
17
|
Chidlow G, Wood JPM, Ebneter A, Casson RJ. Interleukin-6 is an efficacious marker of axonal transport disruption during experimental glaucoma and stimulates neuritogenesis in cultured retinal ganglion cells. Neurobiol Dis 2012; 48:568-81. [PMID: 22884876 DOI: 10.1016/j.nbd.2012.07.026] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Revised: 06/22/2012] [Accepted: 07/25/2012] [Indexed: 01/16/2023] Open
Abstract
It is increasingly recognised that chronically activated glia contribute to the pathology of various neurodegenerative diseases, including glaucoma. One means by which this can occur is through the release of neurotoxic, proinflammatory factors. In the current study, we therefore investigated the spatio-temporal patterns of expression of three such cytokines, IL-1β, TNFα and IL-6, in a validated rat model of experimental glaucoma. First, only weak evidence was found for increased expression of IL-1β and TNFα following induction of ocular hypertension. Second, and much more striking, was that robust evidence was uncovered showing IL-6 to be synthesised by injured retinal ganglion cells following elevation of intraocular pressure and transported in an orthograde fashion along the nerve, accumulating at sites of axonal disruption in the optic nerve head. Verification that IL-6 represents a novel marker of disrupted axonal transport in this model was obtained by performing double labelling immunofluorescence with recognised markers of fast axonal transport. The stimulus for IL-6 synthesis and axonal transport during experimental glaucoma arose from axonal injury rather than ocular hypertension, as the response was identical after optic nerve crush and bilateral occlusion of the carotid arteries, each of which is independent of elevated intraocular pressure. Moreover, the response of IL-6 was not a generalised feature of the gp130 family of cytokines, as it was not mimicked by another family member, ciliary neurotrophic factor. Finally, further study suggested that IL-6 may be an early part of the endogenous regenerative response as the cytokine colocalised with growth-associated membrane phosphoprotein-43 in some putative regenerating axons, and potently stimulated neuritogenesis in retinal ganglion cells in culture, an effect that was additive to that of ciliary neurotrophic factor. These data comprise clear evidence that IL-6 is actively involved in the attempt of injured retinal ganglion cells to regenerate their axons.
Collapse
Affiliation(s)
- Glyn Chidlow
- Ophthalmic Research Laboratories, South Australian Institute of Ophthalmology, Hanson Institute Centre for Neurological Diseases, Frome Rd, Adelaide, SA 5000, Australia.
| | | | | | | |
Collapse
|
18
|
Toops KA, Berlinicke C, Zack DJ, Nickells RW. Hydrocortisone stimulates neurite outgrowth from mouse retinal explants by modulating macroglial activity. Invest Ophthalmol Vis Sci 2012; 53:2046-61. [PMID: 22395888 DOI: 10.1167/iovs.11-8646] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE There is mounting evidence that retinal ganglion cells (RGCs) require a complex milieu of trophic factors to enhance cell survival and axon regeneration after optic nerve injury. The authors' goal was to examine the contribution of components of a combination of hormones, growth factors, steroids, and small molecules to creating a regenerative environment and to determine if any of these components modulated macroglial behavior to aid in regeneration. METHODS Postnatal day 7 mouse retinal explants embedded in collagen were used as an in vitro model of neurite regeneration. Explants were treated with the culture supplements fetal bovine serum, N2, and G5 and a mixture of G5 and N2 components, designated enhanced N2 (EN2). Explants were evaluated for neurite outgrowth over 7 days in culture. The effects of each treatment were also evaluated on cultured RGCs purified by Thy1 immunopanning. Immunohistochemistry and qPCR analysis were used to evaluate differences in gene expression in the explants due to different treatments. RESULTS EN2 stimulated significant neurite outgrowth from explants but not from purified RGCs. Elimination of hydrocortisone (HC) from EN2 reduced the mean neurites per explant by 37%. EN2-treated explants demonstrated increased expression of Gfap, Glul, Glt1, Cntf, Pedf, and VegfA compared with explants treated with EN2 without HC. Subsequent experiments showed that increased expression of Cntf and Glul was critical to the trophic effect of HC. CONCLUSIONS These data suggest that the HC in EN2 indirectly contributed to neurite outgrowth by activating macroglia to produce neurotrophic and neuroprotective molecules.
Collapse
Affiliation(s)
- Kimberly A Toops
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | | | | |
Collapse
|
19
|
The molecular basis of retinal ganglion cell death in glaucoma. Prog Retin Eye Res 2012; 31:152-81. [DOI: 10.1016/j.preteyeres.2011.11.002] [Citation(s) in RCA: 565] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Revised: 10/28/2011] [Accepted: 11/01/2011] [Indexed: 12/14/2022]
|
20
|
Chidlow G, Daymon M, Wood JPM, Casson RJ. Localization of a wide-ranging panel of antigens in the rat retina by immunohistochemistry: comparison of Davidson's solution and formalin as fixatives. J Histochem Cytochem 2011; 59:884-98. [PMID: 21832149 DOI: 10.1369/0022155411418115] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The preferred fixative for whole eyes is Davidson's solution, which provides optimal tissue preservation while avoiding retinal detachment. Hitherto, the compatibility of Davidson's solution with immunohistochemistry has been largely untested. The goal of the present study was to compare the immunolabeling patterns of a wide-ranging panel of commercially available, previously validated antibodies in formalin- and Davidson's-fixed retinas. Immunohistochemistry was performed in normal pigmented rat eyes and, to facilitate localization of inducible proteins, eyes injected with the bacterial toxin lipopolysaccharide or subjected to laser-induced photoreceptor damage. Specificity of labeling was judged by the morphology and distribution of immunopositive cells, by the absence of signal in appropriate controls, and by comparison with expected staining patterns. Retinas fixed in formalin displayed only adequate morphological integrity but were highly compatible with all 39 antibodies evaluated. Retinas fixed in Davidson's solution displayed morphological integrity superior to those fixed in formalin. Generally, the cellular and subcellular patterns and intensities of immunoreactivities obtained with each fixative were identical; however, Davidson's fixative was less compatible with certain antibodies, such as the neurotransmitter γ-aminobutyric acid, the microglial marker iba1, the macroglial stress protein nestin, and the small heat shock proteins Hsp27 and αB-crystallin, shortfalls that somewhat temper enthusiasm concerning its use.
Collapse
Affiliation(s)
- Glyn Chidlow
- Ophthalmic Research Laboratories, South Australian Institute of Ophthalmology, Hanson Institute Centre for Neurological Diseases, Adelaide, Australia.
| | | | | | | |
Collapse
|
21
|
Exogenous modulation of intrinsic optic nerve neuroprotective activity. Graefes Arch Clin Exp Ophthalmol 2010; 248:1105-16. [PMID: 20229104 DOI: 10.1007/s00417-010-1336-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Revised: 02/06/2010] [Accepted: 02/14/2010] [Indexed: 10/19/2022] Open
Abstract
BACKGROUND To characterize the molecular and functional status of the rat retina and optic nerve after acute elevation of intraocular pressure (IOP). METHODS Retinal ischemia was induced in rats by increasing the IOP (110 mmHg/60 minutes). Microarray analysis, quantitative RT-PCR (qRT-PCR) and immunohistochemistry were used to characterize retinal tissue. PLGA microspheres containing neurotrophic factors (BDNF, GDNF, or CNTF) or empty microspheres were injected into the vitreous of operated animals 1 day after elevation of IOP. Pupil light reflex (PLR) parameters and electroretinograms (ERG) were monitored at multiple time points during the 60-day postoperative recovery period. RESULTS Molecular analysis showed a significant intrinsic up-regulation of CNTF at 10 and 25 days after induction of the acute ocular hypertension (p = 0.0067). Molecular tissue analysis of GDNF and its receptors (GDNFR1, GDNFR2), and BDNF and its receptor (trkB) showed no change in expression. Animals that received CNTF microspheres had no significant functional recovery compared to animals which received blank microspheres (p > 0.05). Animals that received GDNF or BDNF microspheres showed significant PLR recovery (p < 0.05 and p < 0.001 respectively) compared to non-treated animals. CONCLUSIONS Continuous release of neurotrophic growth factors (NGFs) significantly protects optic nerve function in the experimental model of retinal ischemia observed by PLR analysis.
Collapse
|
22
|
Bringmann A, Iandiev I, Pannicke T, Wurm A, Hollborn M, Wiedemann P, Osborne NN, Reichenbach A. Cellular signaling and factors involved in Müller cell gliosis: neuroprotective and detrimental effects. Prog Retin Eye Res 2009; 28:423-51. [PMID: 19660572 DOI: 10.1016/j.preteyeres.2009.07.001] [Citation(s) in RCA: 535] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Müller cells are active players in normal retinal function and in virtually all forms of retinal injury and disease. Reactive Müller cells protect the tissue from further damage and preserve tissue function by the release of antioxidants and neurotrophic factors, and may contribute to retinal regeneration by the generation of neural progenitor/stem cells. However, Müller cell gliosis can also contribute to neurodegeneration and impedes regenerative processes in the retinal tissue by the formation of glial scars. This article provides an overview of the neuroprotective and detrimental effects of Müller cell gliosis, with accounts on the cellular signal transduction mechanisms and factors which are implicated in Müller cell-mediated neuroprotection, immunomodulation, regulation of Müller cell proliferation, upregulation of intermediate filaments, glial scar formation, and the generation of neural progenitor/stem cells. A proper understanding of the signaling mechanisms implicated in gliotic alterations of Müller cells is essential for the development of efficient therapeutic strategies that increase the supportive/protective and decrease the destructive roles of gliosis.
Collapse
Affiliation(s)
- Andreas Bringmann
- Department of Ophthalmology and Eye Hospital, University of Leipzig, Liebigstrasse 10-14, D-04103 Leipzig, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
PEDF and GDNF are key regulators of photoreceptor development and retinal neurogenesis in reaggregates from chick embryonic retina. J Ocul Biol Dis Infor 2009; 2:1-11. [PMID: 20072641 PMCID: PMC2802504 DOI: 10.1007/s12177-009-9014-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2008] [Accepted: 01/12/2009] [Indexed: 10/26/2022] Open
Abstract
Here, role(s) of pigment epithelial-derived factor (PEDF) and glial-derived neurotrophic factor (GDNF) on photoreceptor development in three-dimensional reaggregates from the retinae of the E6 chick embryo (rosetted spheroids) was investigated. Fully dispersed cells were reaggregated under serum-reduced conditions and supplemented with 50 ng/ml PEDF alone or in combination with 50 ng/ml GDNF. The spheroids were analyzed for cell growth, differentiation, and death using proliferating cell nuclear antigen, terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick end labeling, and other immunocytochemical stainings and semi-quantitative reverse transcription polymerase chain reaction (RT-PCR) methods. PEDF strongly promoted synthesis of the messenger RNAs for blue and violet cone opsins and to a lesser extent on the red and green cone opsins. This correlated with an increase in the number of cone photoreceptors, as determined by the cone cell marker CERN906. Likewise, PEDF nearly completely inhibited rod differentiation, as detected by immunostaining with anti-rho4D2 and RT-PCR. Furthermore, PEDF accelerated proliferation of cells in the spheroids and inhibited apoptosis. As negative effects, PEDF inhibited the normal histotypic tissue formation of retinal aggregates and reduced the frequency of photoreceptor rosettes and IPL-like areas. Noticeably, supplementation of PEDF-treated cultures with GDNF reversed the effects of PEDF on spheroid morphology and on rod differentiation. This study establishes that PEDF strongly affects three-dimensional retinogenesis in vitro, most notably by inhibiting rod development and supporting proliferation and differentiation of cones, effects which are partially counteracted by GDNF.
Collapse
|
24
|
Ciliary Neurotrophic Factor-Mediated Signaling Regulates Neuronal Versus Glial Differentiation of Retinal Stem Cells/Progenitors by Concentration-Dependent Recruitment of Mitogen-Activated Protein Kinase and Janus Kinase-Signal Transducer and Activator of. Stem Cells 2008; 26:2611-24. [DOI: 10.1634/stemcells.2008-0222] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
25
|
Müller A, Hauk TG, Fischer D. Astrocyte-derived CNTF switches mature RGCs to a regenerative state following inflammatory stimulation. ACTA ACUST UNITED AC 2007; 130:3308-20. [PMID: 17971355 DOI: 10.1093/brain/awm257] [Citation(s) in RCA: 189] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Retinal ganglion cells (RGCs) normally fail to regenerate injured axons and undergo apoptosis soon after injury. We have recently shown that lens injury (LI) or intravitreally applied zymosan allow RGCs to survive axotomy and regenerate axons in the injured optic nerve. Activated macrophages and oncomodulin have been suggested to be the principal mediators of this phenomenon. However, several lines of evidence show that macrophage-derived factors alone cannot account for all the beneficial effects of intraocular inflammation. We show here that LI or zymosan induce upregulation of ciliary neurotrophic factor (CNTF) in retinal astrocytes and release CNTF independent of macrophages and activate the transcription factor signal transducers and activators of transcription 3 (STAT3) in RGCs. Levels of CNTF expressed in retinal glia and STAT3 activation in RGC were correlated with the time course of RGCs switching to an active regenerative state. Intravitreal injections of antibodies against CNTF or a Janus-kinase inhibitor compromised the beneficial effects of LI, whereas an antiserum against oncomodulin was ineffective. Like the action of CNTF, the effects of LI were potentiated by drugs that increase intracellular cAMP levels, resulting in strong axon regeneration in vivo. These data indicate that astrocyte-derived CNTF is a major contributor to the neuroprotective and axon-growth-promoting effects of LI and zymosan. These findings could lead to the development of a therapeutic principle for promoting axon regeneration in the CNS as a whole.
Collapse
Affiliation(s)
- Adrienne Müller
- Department of Experimental Neurology, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | | | | |
Collapse
|
26
|
Expression of ciliary neurotrophic factor after induction of ocular hypertension in the retina of rats. Chin Med J (Engl) 2007. [DOI: 10.1097/00029330-200710020-00019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
27
|
|
28
|
Lönngren U, Näpänkangas U, Lafuente M, Mayor S, Lindqvist N, Vidal-Sanz M, Hallböök F. The growth factor response in ischemic rat retina and superior colliculus after brimonidine pre-treatment. Brain Res Bull 2006; 71:208-18. [PMID: 17113948 DOI: 10.1016/j.brainresbull.2006.09.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2006] [Revised: 08/08/2006] [Accepted: 09/06/2006] [Indexed: 10/24/2022]
Abstract
The alpha-2-adrenergic receptor agonist brimonidine has been shown to increase survival of retinal ganglion cells following ischemic injury to the rat retina. Increased expression of growth factors has been suggested to be involved in this action. We investigated expressional changes of growth factors and their receptors following transient retinal ischemia induced by selective ligature of ophthalmic vessels in rats pre-treated with vehicle or 0.5% brimonidine. In addition, analysis of expression in retinal samples following unilateral administration of brimonidine to normal tissue was performed. Tissue samples of retina and superior colliculus were collected at time points between 6h and 14 days of retinal reperfusion. Analysis of mRNA levels of the ligands BDNF, NT3, CNTF, FGF1, FGF2, FGF9 and HGF; as well as the receptors TrkB, TrkC, p75(NTR), CNTFRalpha, FGFR1, FGFR3, FGFR4 and HGFR were performed using qRT-PCR. The cell specific markers Thy1 and GFAP were analysed. We report transiently increased retinal levels of BDNF, NT3, p75(NTR), FGFR1 and HGFR and decreased levels of FGF9, HGF, TrkB, TrkC, FGFR4 and Thy1 following ischemia. The decreases were counteracted by brimonidine. Brimonidine treatment gave an increase in BDNF, NT3 and CNTF levels compared to the vehicle treated group. In superior colliculus increased levels of growth factor mRNA were found. In conclusion, transient ischemia has a profound effect on gene expression in rat retina. Alterations can also be seen in the superior colliculus but are smaller. Brimonidine pre-treatment attenuates an acute injury-induced response by decreasing the expression of several genes, among them p75(NTR). Brimonidine also causes a prolonged increase of several growth factors as well as receptors in retina and superior colliculus compared to the ischemic situation. The increased expression of several growth factors represents a coordinated growth factor system response that differs from the ischemia-induced changes and is likely part of the neuroprotective activity that is elicited by BMD pre-treatment.
Collapse
Affiliation(s)
- Ulrika Lönngren
- Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | | | | | | | | | | | | |
Collapse
|
29
|
Harvey AR, Hu Y, Leaver SG, Mellough CB, Park K, Verhaagen J, Plant GW, Cui Q. Gene therapy and transplantation in CNS repair: The visual system. Prog Retin Eye Res 2006; 25:449-89. [PMID: 16963308 DOI: 10.1016/j.preteyeres.2006.07.002] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Normal visual function in humans is compromised by a range of inherited and acquired degenerative conditions, many of which affect photoreceptors and/or retinal pigment epithelium. As a consequence the majority of experimental gene- and cell-based therapies are aimed at rescuing or replacing these cells. We provide a brief overview of these studies, but the major focus of this review is on the inner retina, in particular how gene therapy and transplantation can improve the viability and regenerative capacity of retinal ganglion cells (RGCs). Such studies are relevant to the development of new treatments for ocular conditions that cause RGC loss or dysfunction, for example glaucoma, diabetes, ischaemia, and various inflammatory and neurodegenerative diseases. However, RGCs and associated central visual pathways also serve as an excellent experimental model of the adult central nervous system (CNS) in which it is possible to study the molecular and cellular mechanisms associated with neuroprotection and axonal regeneration after neurotrauma. In this review we present the current state of knowledge pertaining to RGC responses to injury, neurotrophic and gene therapy strategies aimed at promoting RGC survival, and how best to promote the regeneration of RGC axons after optic nerve or optic tract injury. We also describe transplantation methods being used in attempts to replace lost RGCs or encourage the regrowth of RGC axons back into visual centres in the brain via peripheral nerve bridges. Cooperative approaches including novel combinations of transplantation, gene therapy and pharmacotherapy are discussed. Finally, we consider a number of caveats and future directions, such as problems associated with compensatory sprouting and the reformation of visuotopic maps, the need to develop efficient, regulatable viral vectors, and the need to develop different but sequential strategies that target the cell body and/or the growth cone at appropriate times during the repair process.
Collapse
Affiliation(s)
- Alan R Harvey
- School of Anatomy and Human Biology, The University of Western Australia, Crawley, WA 6009, Australia
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Mawatari Y, Fukushima M, Inoue T, Setoguchi T, Taga T, Tanihara H. Preferential differentiation of neural progenitor cells into the glial lineage through gp130 signaling in N-methyl-d-aspartate-treated retinas. Brain Res 2005; 1055:7-14. [PMID: 16098488 DOI: 10.1016/j.brainres.2005.06.047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2005] [Revised: 05/31/2005] [Accepted: 06/05/2005] [Indexed: 11/24/2022]
Abstract
The purpose of this study was to investigate the differentiation of neural progenitor cells (NPCs) following retinal transplantation in N-methyl-D-aspartate (NMDA)-treated eyes. NMDA was injected into the vitreous cavity of adult rat eyes. NPCs were prepared from telencephalic neuroepithelium of enhanced green fluorescence protein (EGFP) transgenic mice on embryonic day 14.5. A cell suspension was injected into the vitreous cavity in experimental eyes. Immunohistochemistry was conducted at 1, 2 or 4 weeks after transplantation of NPCs in an effort to determine the survival and differentiation of transplanted NPCs. Similar experiments were conducted using glycoprotein (gp)130-null (-/-) mice. Examination of retinal sections revealed that transplanted NPCs could survive for at least 4 weeks in NMDA-treated retinas. Immunohistochemical studies for specific cell-type markers revealed that, among the transplanted NPCs at 2 weeks after transplantation, the mean percentage (+/-standard deviation) of glial fibrillary acidic protein (GFAP)-positive (glial) cells was 63.5 +/- 7.4%, demonstrating the differentiation of transplanted NPCs with a preference for the glial lineage. Furthermore, the mean percentage of betaIII-tubulin-positive (mature neuronal) cells was 18.8 +/- 4.5%. Following transplantation of NPCs isolated from gp130-/- mice into NMDA-treated retinas, the mean percentage of GFAP-positive cells (17.6 +/- 7.0%), was significantly lower than that in NPCs isolated from wild-type mice (59.1 +/- 6.0%, P = 0.04, Mann-Whitney U test). Preferential differentiation of NPCs into the glial lineage is induced through gp130 signaling in NMDA-treated eyes.
Collapse
Affiliation(s)
- Yuki Mawatari
- Department of Ophthalmology and Visual Science, Kumamoto University Graduate School of Medical Sciences, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | | | | | | | | | | |
Collapse
|
31
|
Munemasa Y, Ohtani-Kaneko R, Kitaoka Y, Kuribayashi K, Isenoumi K, Kogo J, Yamashita K, Kumai T, Kobayashi S, Hirata K, Ueno S. Contribution of mitogen-activated protein kinases to NMDA-induced neurotoxicity in the rat retina. Brain Res 2005; 1044:227-40. [PMID: 15885221 DOI: 10.1016/j.brainres.2005.03.014] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2004] [Revised: 02/24/2005] [Accepted: 03/01/2005] [Indexed: 01/09/2023]
Abstract
We examined the contributions of the mitogen-activated protein kinases (MAPKs) family [extracellular signal-regulated kinase (ERK), p38 kinase (p38), and c-Jun N-terminal kinase (JNK)] to N-methyl-D-aspartate (NMDA)-induced neurotoxicity in the rat retina. Detection of apoptotic cell death in the retinal ganglion cell layer (RGCL) and the inner nuclear layer (INL) by terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick-end labeling (TUNEL) staining began 6 h after intravitreal NMDA (100 nmol) injection and continued to increase thereafter. Western blot analysis showed that phosphorylated MAPKs (p-MAPKs) were expressed in the retina following a temporal manner: maximal expression of phosphorylated ERK (p-ERK) at 1 h, maximal expression of phosphorylated p38 (p-p38) at 6 h, and beginning of phosphorylated JNK (p-JNK) significant increase at 6 h after injection. An immunohistochemical/TUNEL co-localization study showed that p-JNK- and p-p38-positive cells in the RGCL were frequently TUNEL-positive, whereas few p-ERK-positive cells were TUNEL-positive. Moreover, co-injection of inhibitors for JNK (0.2 nmol SP600125) and/or p38 (2.0 nmol SB203580) with NMDA was effective in ameliorating NMDA-induced apoptotic cell loss in the RGCL 12 h after injection, as shown by TUNEL-positive cell counts. These inhibitors also protected the inner retina as shown by morphometric studies such as cell counts in the RGCL and measurement of the IPL thickness 7 days after injection. On the other hand, an ERK inhibitor (2.0 nmol U0126) did not suppress NMDA-induced cell death in the RGCL nor thinning of the IPL. These findings suggest that JNK and p38 are proapoptotic in NMDA-induced cell death in the RGCL, but not ERK.
Collapse
Affiliation(s)
- Yasunari Munemasa
- Department of Ophthalmology, Saint Marianna University School of Medicine, 2-16-1 Sugao, Kawasaki-shi, Kanagawa 216-8511, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Maier K, Rau CR, Storch MK, Sättler MB, Demmer I, Weissert R, Taheri N, Kuhnert AV, Bähr M, Diem R. Ciliary neurotrophic factor protects retinal ganglion cells from secondary cell death during acute autoimmune optic neuritis in rats. Brain Pathol 2005; 14:378-87. [PMID: 15605985 PMCID: PMC8095794 DOI: 10.1111/j.1750-3639.2004.tb00081.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the CNS which leads to demyelination, axonal destruction and neuronal loss in the early stages. Available therapies mainly target the inflammatory component of the disease but fail to prevent neurodegeneration. To investigate the effect of ciliary neurotrophic factor (CNTF) on the survival of retinal ganglion cells (RGCs), the neurons that form the axons of the optic nerve, we used a rat model of myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis. Optic neuritis in this model was diagnosed by recording visual evoked potentials, and RGC function was monitored by measuring electroretinograms. This study demonstrates that CNTF has a neuroprotective effect on affected RGCs during acute optic neuritis. Furthermore, we demonstrate that CNTF exerts its neuroprotective effect through activation of the Janus kinase/signal transducer and activator of transcription pathway, mitogen activated protein kinases and a shift in the Bcl-2 family of proteins towards the anti-apoptotic side. In summary, our results demonstrate that CNTF can serve as an effective neuroprotective treatment in a rat model of MS that especially reflects the neurodegenerative aspects of this disease.
Collapse
|
33
|
Huang SP, Lin PK, Liu JH, Khor CN, Lee YJ. Intraocular gene transfer of ciliary neurotrophic factor rescues photoreceptor degeneration in RCS rats. J Biomed Sci 2004; 11:37-48. [PMID: 14730208 DOI: 10.1007/bf02256547] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2003] [Accepted: 09/01/2003] [Indexed: 12/01/2022] Open
Abstract
Ciliary neurotrophic factor (CNTF) is known as an important factor in the regulation of retinal cell growth. We used both recombinant CNTF and an adenovirus carrying the CNTF gene to regulate retinal photoreceptor expression in a retinal degenerative animal, Royal College of Surgeons (RCS) rats. Cells in the outer nuclear layer of the retinae from recombinant-CNTF-treated, adenoviral-CNTF-treated, saline-operated, and contralateral untreated preparations were examined for those exhibiting CNTF photoreceptor protective effects. Cell apoptosis in the outer nuclear layer of the retinae was also detected. It was found that CNTF had a potent effect on delaying the photoreceptor degeneration process in RCS rats. Furthermore, adenovirus CNTF gene transfer was proven to be better at rescuing photoreceptors than that when using recombinant CNTF, since adenoviral CNTF prolonged the photoreceptor protection effect. The function of the photoreceptors was also examined by taking electroretinograms of different animals. Adenoviral-CNTF-treated eyes showed better retinal function than did the contralateral control eyes. This study indicates that adenoviral CNTF effectively rescues degenerating photoreceptors in RCS rats.
Collapse
Affiliation(s)
- Shun-Ping Huang
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | | | | | | | | |
Collapse
|
34
|
Bhattacharya S, Dooley C, Soto F, Madson J, Das AV, Ahmad I. Involvement of Ath3 in CNTF-mediated differentiation of the late retinal progenitors. Mol Cell Neurosci 2004; 27:32-43. [PMID: 15345241 DOI: 10.1016/j.mcn.2004.05.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2003] [Revised: 04/22/2004] [Accepted: 05/10/2004] [Indexed: 11/26/2022] Open
Abstract
The cellular diversity of the mammalian retina is underpinned by multipotential neural progenitors that generate retinal neurons and glia with temporal and spatial specificity. It is thought, based on studies using a variety of approaches, that the fate of retinal progenitors is determined through interactions between temporally and spatially arrayed epigenetic cues with intrinsic factors that regulate the competence of cells to respond to such cues. Here, we demonstrate interactions between an intrinsic factor Ath3, a neural bHLH protein, and an extrinsic factor CNTF during the differentiation of the late retinal progenitors along the bipolar cell lineage. Expression of Ath3 is predominantly associated with the late stage of retinal histogenesis when bipolar cells are specified, and in adult it is detected in cells expressing bipolar cell-specific markers. We demonstrate that CNTF-induced bipolar cell differentiation is accompanied by an increase in levels of Ath3 transcripts and compromised when Ath3 expression is attenuated. Our study suggests that the influence of CNTF on the differentiation of late retinal progenitors is mediated through Ath3.
Collapse
MESH Headings
- Amino Acid Sequence/genetics
- Animals
- Animals, Newborn
- Base Sequence/genetics
- Basic Helix-Loop-Helix Transcription Factors
- Biomarkers
- Cell Differentiation/drug effects
- Cell Differentiation/genetics
- Ciliary Neurotrophic Factor/metabolism
- Ciliary Neurotrophic Factor/pharmacology
- DNA, Complementary/analysis
- DNA, Complementary/genetics
- Eye Proteins/genetics
- Eye Proteins/isolation & purification
- Eye Proteins/metabolism
- Gene Expression Regulation, Developmental/drug effects
- Gene Expression Regulation, Developmental/genetics
- Helix-Loop-Helix Motifs/genetics
- Molecular Sequence Data
- Nerve Tissue Proteins/drug effects
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/isolation & purification
- Nerve Tissue Proteins/metabolism
- Neuroglia/cytology
- Neuroglia/metabolism
- Neurons/cytology
- Neurons/drug effects
- Neurons/metabolism
- Photoreceptor Cells, Vertebrate/cytology
- Photoreceptor Cells, Vertebrate/drug effects
- Photoreceptor Cells, Vertebrate/metabolism
- RNA, Messenger/drug effects
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Retina/cytology
- Retina/growth & development
- Retina/metabolism
- Stem Cells/cytology
- Stem Cells/drug effects
- Stem Cells/metabolism
Collapse
Affiliation(s)
- Sumitra Bhattacharya
- Department Ophthalmology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | | | | | | | | | | |
Collapse
|
35
|
Thanos S, Naskar R. Correlation between retinal ganglion cell death and chronically developing inherited glaucoma in a new rat mutant. Exp Eye Res 2004; 79:119-29. [PMID: 15183107 DOI: 10.1016/j.exer.2004.02.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2003] [Accepted: 02/04/2004] [Indexed: 11/20/2022]
Abstract
Glaucoma is a progressive optic neuropathy with characteristic optic disc changes, retinal ganglion cell loss and progressive visual field defects. Elevated intraocular pressure is considered to be a major risk factor in glaucomatous neuropathy. This study aimed to characterize and document a new chronic glaucoma model in the rat with respect to the effect of elevated intraocular pressure on overall retinal dysfunction and retinal ganglion cell loss, and to elucidate the possible mechanisms underlying this cell loss. Intraocular pressure (IOP) was measured in rats using a Tonopen. RGCs were retrogradely labeled with the fluorescent dye, 4-[didecylaminostyryl]-N-methyl-pyridinium-iodide (4-Di-10 ASP) and quantified on retinal flat mounts using fluorescence microscopy. The optic nerve head was examined fundoscopically. Changes in the histological appearance of the whole eyes was studied in paraffin sections, and immunohistochemistry was carried out on cryostat sections. The levels of mRNA for several genes were compared between control and glaucomatous retinae using semi-quantitative RT-PCR. Mutant animals are affected with either a unilateral or bilateral enlargement of the globes having an IOP that ranged from 25 to 45 mmHg, as compared to control values of 12-16 mmHg. The IOP of glaucomatous eyes increased significantly with age to attain a value of 35+/-7.3 at 1.5 years. Concomitant with the rise in IOP, the number of labeled RGCs continued to decrease in number with age. A total of 1887+/-117RGC mm(-2) could be labeled in wild-type control and juvenile mutant pre-glaucomatous retinas, whereas this number dropped to 92+/-26RGC mm(-2) at 1.5 years. Ophthalmoscopy revealed atrophied optic nerve heads in the affected eyes. The pars plicata and the pars plana of the ciliary body of glaucomatous eyes were hypertrophied and elongated, respectively. The anterior chamber was narrow and the irido-corneal angle open in glaucoma eyes. The mRNA of glial-fibrillary-acidic protein, endothelin-1, STAT-3 and STAT-6 increased in the retinas correlating with the severity and duration of the disease. Changes in the expression of GFAP and endothelin-1 could be confirmed using immunohistochemistry. This model may help to address several fundamental issues in the pathogenesis of glaucoma and aid in the development of neuroprotective strategies.
Collapse
Affiliation(s)
- Solon Thanos
- Department of Experimental Ophthalmology, School of Medicine, University Eye Hospital Münster, Domagkstrasse 15, D-48149 Münster, Germany.
| | | |
Collapse
|
36
|
Sarup V, Patil K, Sharma SC. Ciliary neurotrophic factor and its receptors are differentially expressed in the optic nerve transected adult rat retina. Brain Res 2004; 1013:152-8. [PMID: 15193523 DOI: 10.1016/j.brainres.2004.03.030] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2004] [Indexed: 11/23/2022]
Abstract
Ciliary neurotrophic factor (CNTF) has been implicated in the survival and regeneration of adult rat retinal ganglion cells (RGCs). Expression of CNTF and its receptors, ciliary neurotrophic factor receptor alpha (CNTFRalpha) and leukemia inhibitory factor receptor (LIFR) was studied in normal, 6-h, 1-, and 3-day optic nerve transected retinas. Optic nerve transection was performed on the right eye in one group. In the other group, it was performed 5 days after intratectal Fluorogold injection. Retinas were studied using immunohistochemical labeling and Western blot analysis. Astrocytes were labeled with GFAP and Müller cells with glutamine synthetase. CNTF was observed in identifiable astrocytes and Müller cells, and its intensity increased in the experimental retinas. CNTFRalpha was localized on RGCs in experimental retinas. LIFR was localized on RGCs and Müller cells in normal and experimental retinas. CNTF and its receptors are differentially expressed after optic nerve transection and could help delay RGC death in such a stressful environment.
Collapse
Affiliation(s)
- Vimal Sarup
- Department of Ophthalmology and Cell Biology and Anatomy, New York Medical College, Valhalla, NY-10595, USA
| | | | | |
Collapse
|
37
|
Ji JZ, Elyaman W, Yip HK, Lee VWH, Yick LW, Hugon J, So KF. CNTF promotes survival of retinal ganglion cells after induction of ocular hypertension in rats: the possible involvement of STAT3 pathway. Eur J Neurosci 2004; 19:265-72. [PMID: 14725620 DOI: 10.1111/j.0953-816x.2003.03107.x] [Citation(s) in RCA: 141] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We examined the neuroprotective effect of ciliary neurotrophic factor (CNTF) on retinal ganglion cells (RGCs) in a rat glaucoma model with increased intraocular pressure (IOP) and studied the CNTF-mediated activation of Janus kinase/signal transducer and activator of transcription (JAK-STAT) pathway. Elevated IOP was induced by laser photocoagulation of the episcleral and limbal veins. The survival of RGCs was studied using Fluoro-Gold labelled in ocular hypertensive eyes with or without CNTF intravitreal injection. Immunochemical staining and immunoblot analysis for CNTF and phosphorylated STAT3 (pSTAT3) were performed. There was a significant and progressive loss of RGCs in the retinas following the induction of elevated IOP. A single intravitreal injection of 2 microg in 2 microL CNTF significantly protected RGCs up to 4 weeks. pSTAT3 was only transiently expressed in ocular hypertensive eyes. However, in eyes treated with CNTF, pSTAT3 was observed up to 2 weeks after the induction of elevated IOP. In ocular hypertensive eyes, CNTF-positive cells were found in the inner nuclear layer (INL), and there was a transient increase in the pSTAT3 cells in the ganglion cell layer and INL. Immunoblots showed that STAT3 was transiently phosphorylated after IOP increase, but with an injection of CNTF, pSTAT3 protein was observed up to 2 weeks after hypertensive glaucoma induction. Laser-induced chronic ocular hypertension in rats resulted in the death of RGCs and a transient activation of STAT3 in the retina. Intravitreal injection of CNTF showed a significant protection of RGCs, and the JAK-STAT signalling could be one of the important pathways that underlie the mechanism of CNTF neuroprotection in this rat glaucoma model.
Collapse
Affiliation(s)
- Jian-Zhong Ji
- Department of Anatomy, The University of Hong Kong, Hong Kong, China
| | | | | | | | | | | | | |
Collapse
|
38
|
Osborne NN, Casson RJ, Wood JPM, Chidlow G, Graham M, Melena J. Retinal ischemia: mechanisms of damage and potential therapeutic strategies. Prog Retin Eye Res 2004; 23:91-147. [PMID: 14766318 DOI: 10.1016/j.preteyeres.2003.12.001] [Citation(s) in RCA: 753] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Retinal ischemia is a common cause of visual impairment and blindness. At the cellular level, ischemic retinal injury consists of a self-reinforcing destructive cascade involving neuronal depolarisation, calcium influx and oxidative stress initiated by energy failure and increased glutamatergic stimulation. There is a cell-specific sensitivity to ischemic injury which may reflect variability in the balance of excitatory and inhibitory neurotransmitter receptors on a given cell. A number of animal models and analytical techniques have been used to study retinal ischemia, and an increasing number of treatments have been shown to interrupt the "ischemic cascade" and attenuate the detrimental effects of retinal ischemia. Thus far, however, success in the laboratory has not been translated to the clinic. Difficulties with the route of administration, dosage, and adverse effects may render certain experimental treatments clinically unusable. Furthermore, neuroprotection-based treatment strategies for stroke have so far been disappointing. However, compared to the brain, the retina exhibits a remarkable natural resistance to ischemic injury, which may reflect its peculiar metabolism and unique environment. Given the increasing understanding of the events involved in ischemic neuronal injury it is hoped that clinically effective treatments for retinal ischemia will soon be available.
Collapse
Affiliation(s)
- Neville N Osborne
- Nuffield Laboratory of Ophthalmology, University of Oxford, Walton Street, Oxford OX2 6AW, UK.
| | | | | | | | | | | |
Collapse
|
39
|
Nakamichi N, Chidlow G, Osborne NN. Effects of intraocular injection of a low concentration of zinc on the rat retina. Neuropharmacology 2003; 45:637-48. [PMID: 12941377 DOI: 10.1016/s0028-3908(03)00206-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The main aim of this study was to investigate whether intraocular injection of low concentrations of zinc (no greater than 10 microM) aid the survival of ganglion cells in the rat retina after excitotoxic (NMDA) and ischemia/reperfusion injuries. We also determined whether low amounts of zinc cause any detectable retinal toxicity. Intraocular injection of NMDA caused substantial reductions in the mRNA levels of the ganglion cell-specific markers Thy-1 and neurofilament light (NF-L). Co-injection of 0.1 or 1 nmol zinc neither diminished nor exacerbated the effect of NMDA on the levels of these mRNAs. Likewise, ischemia/reperfusion caused significant decreases in the levels of Thy-1 and NF-L mRNAs and in the b-wave amplitude of the electroretinogram. These effects were not counteracted by injection of zinc. Intraocular injection of NMDA caused marked toxicological effects in retinal glial cells, including upregulations of cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), glial fibrial acidic protein (GFAP), basic fibroblast growth factor (FGF-2) and ciliary neurotrophic factor (CNTF). Interestingly, injection of 1 nmol zinc caused no changes in the levels of COX-2 and iNOS, yet produced similar, although quantitatively less pronounced, changes in FGF-2, GFAP and CNTF. The upregulations of FGF-2 and CNTF suggest that increasing zinc intake may benefit injured retinal neurons. However, this was not found to be the case in the present studies, perhaps due to the acute nature of the injury paradigms utilised.
Collapse
Affiliation(s)
- N Nakamichi
- Laboratory of Molecular Pharmacology, Kanazawa University Graduate School of Natural Science and Technology, 13-1 Takara-machi, Kanazawa, Ishikawa 920-0934, Japan
| | | | | |
Collapse
|
40
|
Valter K, Bisti S, Stone J. Location of CNTFRalpha on outer segments: evidence of the site of action of CNTF in rat retina. Brain Res 2003; 985:169-75. [PMID: 12967721 DOI: 10.1016/s0006-8993(03)03130-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ciliary neurotrophic factor (CNTF) is an important factor in the retina's mechanisms of self-protection. It is generated by retinal glial cells in response to stress, and has a significant protective effect on retinal neurones. In this study we have identified the location of the alpha component of the CNTF receptor complex (CNTFRalpha) in rat retina, using immunohistochemistry and high-resolution confocal microscopy. The major location of CNTFRalpha is on photoreceptor outer segments. More scattered, granular forms of CNTFRalpha were identified in association with Müller cell processes in other retinal layers. Colocalisation of CNTF with CNTFRalpha, suggestive of ligand-receptor binding, was detected on outer segments, and in both normal retinas and retinas stressed by light or oxygen. Results provide evidence that the principal site of CNTF action is the outer segments of photoreceptors. This confirms the known ability of CNTF to protect photoreceptors against stress, and suggest that it acts by modulating mechanisms specific to the outer segment, such as the phototransduction cascade or the membrane channels, which control dark current.
Collapse
Affiliation(s)
- Krisztina Valter
- CNS Stability and Degeneration Group, Research School of Biological Sciences, Australian National University, Canberra, ACT 2601, Australia.
| | | | | |
Collapse
|
41
|
Böttcher T, Mix E, Koczan D, Bauer P, Pahnke J, Peters S, Weinelt S, Knoblich R, Strauss U, Cattaneo E, Thiesen HJ, Rolfs A. Gene expression profiling of ciliary neurotrophic factor-overexpressing rat striatal progenitor cells (ST14A) indicates improved stress response during the early stage of differentiation. J Neurosci Res 2003; 73:42-53. [PMID: 12815707 DOI: 10.1002/jnr.10624] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Neuronal progenitor cells delivering neurotrophic factors are a promising therapeutic tool for treatment of neurodegenerative diseases. Although several promising results have come from studies in different animal models, detailed knowledge of the action of neurotrophic factors in the CNS is still lacking. A clonally derived, immortalized rat striatal cell line (ST14A) expressing ciliary neurotrophic factor (CNTF) offers a stable and controlled background with which to analyze CNTF actions on the transcriptional level in CNS progenitor cells. To identify early transcriptional changes induced by CNTF expression, we transfected the CNTF gene into ST14A cells, which differentiate at the nonpermissive temperature of 39 degrees C via suppression of the immortalizing SV40 large T antigen. This shows a CNTF-dependent hypoxic/ischemic stress response during the earliest stage of differentiation, with expression of specific transcripts and evidence of translational repression leading to decreased protein synthesis in the transfected cells. This process is mediated by the Ras/MAP kinase pathway and is accompanied by impaired proliferation and metabolism as well as signs of neuronal differentiation. The stress-like response in the early stage of differentiation improves the ability of the transfected cells to respond to and cope with a stressful environment in vivo. The present data indicate higher viability, longer life, and greater differentiation capacity of CNTF-ST14A cells if they are used for transplantation. We conclude that the stress-like response during the early stage of differentiation improves the ability of the CNTF-ST14A cells to respond and adapt to a stressful environment, which renders them useful candidate cells for in vivo trials of treatment for neurodegenerative diseases in animal models, e.g., of Huntington's disease.
Collapse
Affiliation(s)
- Tobias Böttcher
- Department of Neurology, University of Rostock, Rostock, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
van Adel BA, Kostic C, Déglon N, Ball AK, Arsenijevic Y. Delivery of ciliary neurotrophic factor via lentiviral-mediated transfer protects axotomized retinal ganglion cells for an extended period of time. Hum Gene Ther 2003; 14:103-15. [PMID: 12614562 DOI: 10.1089/104303403321070801] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Ciliary neurotrophic factor (CNTF) has recently been demonstrated to be one of the most promising neurotrophic factors to improve both the survival and regeneration of injured retinal ganglion cells (RGCs). In the present study, we used optic nerve transection as an in vivo model to evaluate the effectiveness of a self-inactivating, replication-deficient lentiviral-mediated transfer of human ciliary neurotrophic factor (SIN-PGK-CNTF) on the survival of axotomized adult rat RGCs. Counts of dextran-fluorescein isothiocyanate conjugated (D-FITC)-retrogradely labeled RGCs revealed that the percentage of RGCs was drastically reduced (<90% cell death) 21 days after optic nerve transection. Retinal sections stained with X-gal revealed that intravitreal injection of the control LacZ-expressing lentiviral vector (LV-LacZ) resulted in the transduction of RGCs and retinal pigment epithelium (RPE) cells. A single intravitreal injection of LV-CNTF at the time of axotomy significantly enhanced RGC survival at 14 and 21 days postaxotomy compared to controls. These results demonstrate for the first time that rapid and prolonged delivery of CNTF using lentiviral-mediated gene transfer to the retina is an effective treatment for rescuing axotomized RGCs for an extended period of time. These results suggest that early and continuous administration of CNTF could serve as a potential treatment for retinal disorders involving optic neuropathy and RGC injury such as in glaucoma.
Collapse
Affiliation(s)
- Brian A van Adel
- Department of Pathology and Molecular Medicine, Faculty of Health Sciences, McMaster University, Hamilton, L8N 3ZS, Canada
| | | | | | | | | |
Collapse
|
43
|
Microglia-Müller glia cell interactions control neurotrophic factor production during light-induced retinal degeneration. J Neurosci 2002. [PMID: 12417648 DOI: 10.1523/jneurosci.22-21-09228.2002] [Citation(s) in RCA: 274] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Activation of microglia commonly occurs in response to a wide variety of pathological stimuli including trauma, axotomy, ischemia, and degeneration in the CNS. In the retina, prolonged or high-intensity exposure to visible light leads to photoreceptor cell apoptosis. In such a light-reared retina, we found that activated microglia invade the degenerating photoreceptor layer and alter expression of neurotrophic factors such as nerve growth factor (NGF), ciliary neurotrophic factor (CNTF), and glial cell line-derived neurotrophic factor (GDNF). Because these neurotrophic factors modulate secondary trophic factor expression in Müller glial cells, microglia-Müller glia cell interaction may contribute to protection of photoreceptors or increase photoreceptor apoptosis. In the present study, we demonstrate the possibility that such functional glia-glia interactions constitute the key mechanism by which microglia-derived NGF, brain-derived neurotrophic factor (BDNF), and CNTF indirectly influence photoreceptor survival, although the receptors for these neurotrophic factors are absent from photoreceptors, by modulating basic fibroblast growth factor (bFGF) and GDNF production and release from Müller glia. These observations suggest that microglia regulate the microglia-Müller glia-photoreceptor network that serves as a trophic factor-controlling system during retinal degeneration.
Collapse
|
44
|
Akiyama H, Nakazawa T, Shimura M, Tomita H, Tamai M. Presence of mitogen-activated protein kinase in retinal Müller cells and its neuroprotective effect ischemia-reperfusion injury. Neuroreport 2002; 13:2103-7. [PMID: 12438934 DOI: 10.1097/00001756-200211150-00022] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The purpose of this study was to determine whether the mitogen-activated protein kinase (MAPK) signaling pathway in the retina plays a neuroprotective role against ischemia- reperfusion injury. Western blot analysis showed that the MAPK activity was markedly increased within an hour after ischemia-reperfusion and subsequently decreased. Immunohistochemical studies revealed that MAPK was expressed mainly in the retinal Müller cells (RMCs). Pre-ischemic intravitreal administration of a MAPK inhibitor, U0126, increased the number of ganglion cell deaths induced by ischemia-reperfusion injury. We conclude that the MAPK activated in the RMCs protects ganglion cells against the ischemia-reperfusion injury through glia-neuronal interaction.
Collapse
Affiliation(s)
- Hiroshi Akiyama
- Department of Ophthalmology, Tohoku University School of Medicine, 1-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8574, Japan
| | | | | | | | | |
Collapse
|
45
|
Chidlow G, Schmidt KG, Wood JPM, Melena J, Osborne NN. Alpha-lipoic acid protects the retina against ischemia-reperfusion. Neuropharmacology 2002; 43:1015-25. [PMID: 12423671 DOI: 10.1016/s0028-3908(02)00129-6] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The aim of this study was to examine whether the antioxidant alpha-lipoic acid protects retinal neurons from ischemia-reperfusion injury. Rats were injected intraperitoneally with either vehicle or alpha-lipoic acid (100 mg/kg) once daily for 11 days. On the third day, ischemia was delivered to the rat retina by raising the intraocular pressure above systolic blood pressure for 45 min. The electroretinogram was measured prior to ischemia and 5 days after reperfusion. Rats were killed 5 or 8 days after reperfusion and the retinas were processed for immunohistochemistry and for determination of mRNA levels by RT-PCR. Ischemia-reperfusion caused a significant reduction of the a- and b-wave amplitudes of the electroretinogram, a decrease in nitric oxide synthase and Thy-1 immunoreactivities, a decrease of retinal ganglion cell-specific mRNAs and an increase in bFGF and CNTF mRNA levels. All of these changes were clearly counteracted by alpha-lipoic acid. Moreover, in mixed rat retinal cultures, alpha-lipoic acid partially counteracted the loss of GABA-immunoreactive neurons induced by anoxia. The results of the study demonstrate that alpha-lipoic acid provides protection to the retina as a whole, and to ganglion cells in particular, from ischemia-reperfusion injuries. alpha-Lipoic acid also displayed negligible affinity for voltage-dependent sodium and calcium channels.
Collapse
Affiliation(s)
- G Chidlow
- Nuffield Laboratory of Ophthalmology, University of Oxford, Walton Street, Oxford OX2 6AW, UK
| | | | | | | | | |
Collapse
|
46
|
Koeberle PD, Ball AK. Neurturin enhances the survival of axotomized retinal ganglion cells in vivo: combined effects with glial cell line-derived neurotrophic factor and brain-derived neurotrophic factor. Neuroscience 2002; 110:555-67. [PMID: 11906793 DOI: 10.1016/s0306-4522(01)00557-7] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In the present study we localized glial cell line-derived neurotrophic factor (GDNF), and the high affinity receptor for GDNF (GFRalpha-1) in the rat retina. We also examined the effects of neurturin on the survival of axotomized retinal ganglion cells (RGCs) and compared neurturin-mediated RGC rescue to GDNF and brain-derived neurotrophic factor (BDNF) neuroprotection. We administered combined injections of neurturin with BDNF or GDNF in order to determine if these factors rescue RGCs by different mechanisms. GDNF immunoreactivity was localized to RGCs, photoreceptors, and retinal pigment epithelial cells. GFRalpha-1 immunoreactivity was localized to RGCs, Müller cells, and photoreceptors. RGC densities in control retinas decreased from the original value of 2481+/-121 (RGCs/mm(2)+/-S.D.) to 347+/-100 at 14 days post-axotomy. Neurturin treatment significantly increased RGC survival after axotomy (745+/-94) similar to GDNF (868+/-110). BDNF treatment resulted in higher RGC survival (1109+/-156) than either neurturin or GDNF. Combined administration of neurturin with BDNF had additive effects on the survival of axotomized RGCs (1962+/-282), similar to combined administration of GDNF and BDNF (1825+/-269). Combined administration of neurturin and GDNF (1265+/-178) had an enhanced effect on RGC survival. These results suggest that neurturin, GDNF, and BDNF act independently to rescue injured RGCs. Our results also suggest that RGCs and retinal Müller cells may be responsive to GDNF because they both express GFRalpha-1. The present findings have implications for the rescue of injured retinal ganglion cells, as well as other CNS neurons that are responsive to neurturin, GDNF, and BDNF, including midbrain dopaminergic neurons and motor neurons.
Collapse
Affiliation(s)
- Paulo D Koeberle
- McMaster University, Anatomy, HSC 1R1, 1200 Main Strreet West, L8N 3Z5, Hamilton, ON, Canada
| | | |
Collapse
|
47
|
Chen H, Weber AJ. Expression of glial fibrillary acidic protein and glutamine synthetase by Müller cells after optic nerve damage and intravitreal application of brain-derived neurotrophic factor. Glia 2002; 38:115-25. [PMID: 11948805 DOI: 10.1002/glia.10061] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Müller glia play an important role in maintaining retinal homeostasis, and brain-derived neurotrophic factor (BDNF) has proven to be an effective retinal ganglion cell (RGC) neuroprotectant following optic nerve injury. The goal of these studies was to investigate the relation between optic nerve injury and Müller cell activation, and to determine the extent to which BDNF affects the injury response of Müller cells. Using immunocytochemistry and Western blot analysis, temporal changes in the expression of glial fibrillary acidic protein (GFAP) and glutamine synthetase (GS) were examined in rats after optic nerve crush alone, or in conjunction with an intravitreal injection of BDNF (5 microg). GFAP protein levels were normal at 1 day post-crush, but increased approximately 9-fold by day 3 and remained elevated over the 2-week period studied. Müller cell GS expression remained stable after optic nerve crush, but the protein showed a transient shift in its cellular distribution; during the initial 24-h period post-crush the GS protein appeared to translocate from the cell body to the inner and outer glial processes, and particularly to the basal endfeet located in the ganglion cell layer. BDNF alone, or in combination with optic nerve crush, did not have a significant effect on the expression of either GFAP or GS compared with the normal retina, or after optic nerve crush alone, respectively. The data indicate that although BDNF is a potent neuroprotectant in the vertebrate retina, it does not appear to have a significant influence on Müller cell expression of either GS or GFAP in response to optic nerve injury.
Collapse
Affiliation(s)
- Hao Chen
- Department of Pharmacology, University of Tennessee at Memphis, USA
| | | |
Collapse
|
48
|
Wahlin KJ, Adler R, Zack DJ, Campochiaro PA. Neurotrophic signaling in normal and degenerating rodent retinas. Exp Eye Res 2001; 73:693-701. [PMID: 11747369 DOI: 10.1006/exer.2001.1078] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Several types of insult cause up-regulation of neurotrophic factors and their receptors in the retina resulting in decreased photoreceptor cell death from subsequent injury. This phenomenon is more prominent in rats than in mice and neurotrophic factors are more efficacious in rats than mice. If up-regulation of neurotrophic factor receptors on photoreceptor cells early in the course of degenerations contributes to neurotrophic factor survival-promoting activity, it may also increase the ability to detect neurotrophic factor-induced signaling in photoreceptors, particularly in rats. In this study, these hypotheses were investigated by performing immunohistochemical staining for the phosphorylated form of extracellular receptor kinase (pERK) or c-fos after intravitreous injection of neurotrophic factors in wild type rats or mice, or those with inherited retinal degenerations. In both rats and mice either early or late in the course of degeneration, or in wild type animals, intravitreous injection of brain-derived neurotrophic factor, ciliary neurotrophic factor, or fibroblast growth factor-2 caused immunostaining for pERK and c-fos in cells of the inner retina, particularly Müller cells, but not in photoreceptors. These data add to the mounting evidence suggesting that neurotrophic factors act indirectly through Müller cells to promote photoreceptor survival.
Collapse
Affiliation(s)
- K J Wahlin
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287-9277, USA
| | | | | | | |
Collapse
|
49
|
Hermann DM, Kilic E, Kügler S, Isenmann S, Bähr M. Adenovirus-mediated GDNF and CNTF pretreatment protects against striatal injury following transient middle cerebral artery occlusion in mice. Neurobiol Dis 2001; 8:655-66. [PMID: 11493030 DOI: 10.1006/nbdi.2001.0399] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
During the last few years, adenoviral gene transfer techniques have achieved increasing interest in the treatment of neurodegenerative diseases. However, gene therapy requires that delivered genes are translated into proteins. This may pose a problem in focal ischemia where protein synthesis is compromized. The present study was conducted to find out the feasibility of adenoviral GDNF and CNTF delivery in transient focal ischemia, as induced by 30 min of intraluminar middle cerebral artery (MCA) occlusion in mice. Injections of vehicle, of an adenoviral vector deleted in the E1 region (Ad-dE1) and of vectors expressing the GDNF (Ad-GDNF), CNTF (Ad-CNTF), or GFP (Ad-EGFP) gene from a CMV promoter were stereotactically placed in the dorsolateral striatum, i.e., the core of the MCA territory, and focal ischemia was induced seven days later. Thread occlusion resulted in disseminated injury of the striatum, but not the overlying cortex. The number of viable neurons was significantly increased after 1 and 3 days of reperfusion both in Ad-GDNF and Ad-CNTF as compared with vehicle or Ad-dE1-treated animals, whereas the number of injured cells was significantly reduced, as shown by cresyl violet staining, terminal transferase biotinylated-dUTP nick end-labeling (TUNEL), and immunocytochemistry for activated caspase-3. Interestingly, the protective effects of Ad-GDNF were similarly strong in areas of the striatum adjacent and remote of the adenoviral infusion site, while Ad-CNTF showed pronounced rescue effects in the surrounding, but rather little effects distant to the infusion. The present study demonstrates that adenoviral delivery of neurotrophic factors may be a useful tool for the treatment of focal ischemia.
Collapse
Affiliation(s)
- D M Hermann
- Department of Neurology, University of Tübingen, Hoppe-Seyler-Strasse 3, D-72076 Tübingen, Germany
| | | | | | | | | |
Collapse
|
50
|
Walsh N, Valter K, Stone J. Cellular and subcellular patterns of expression of bFGF and CNTF in the normal and light stressed adult rat retina. Exp Eye Res 2001; 72:495-501. [PMID: 11311041 DOI: 10.1006/exer.2000.0984] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This study compared the distributions in the normal and light stressed rat retina of the neuroprotective factors bFGF (basic fibroblast growth factor) and CNTF (ciliary neurotrophic factor). Albino Sprague-Dawley rats were raised in cyclic light and some were exposed to bright continuous light for 48 hr, to induce light damage of photoreceptors. Their retinas were prepared as cryosections, immunolabelled with antibodies to bFGF and CNTF and analysed by confocal microscopy. Both factors were prominent in macroglial cells (astrocytes, Müller cells) and the retinal pigment epithelium (RPE). In the somas of these cells the distributions of the two factors were complementary, with bFGF concentrated in the nuclei and CNTF in the cytoplasm. Both factors were distributed along the processes of macroglial cells, in granular form. CNTF was not detected in neurones, but bFGF was consistently present in the cytoplasm of ganglion cell somas and, in regions of retina subject to stress, in the cytoplasm of photoreceptors. bFGF was not detected in the nuclei or processes of neurones. In retina stressed by light exposure or proximity to the anterior edge of the retina, the levels of bFGF and CNTF were up-regulated, without major changes in localization. Macroglial cells (Müller cells, astrocytes) play a major role in distributing bFGF and CNTF throughout the retina. The different localizations of the two factors within the somas of macroglial, RPE and photoreceptor cells, suggest that their protective actions are exerted by distinctive mechanisms.
Collapse
Affiliation(s)
- N Walsh
- NSW Retinal Dystrophy Research Centre, Department of Anatomy and Histology, University of Sydney, NSW 2006, Australia
| | | | | |
Collapse
|