1
|
Gallego-Ortega A, Galindo-Romero C, Vidal-Villegas B, Bernal-Garro JM, de la Villa P, Avilés-Trigueros M, Vidal-Sanz M. The action of 7,8-dihydroxyflavone preserves retinal ganglion cell survival and visual function via the TrkB pathway in NMDA-induced retinal excitotoxicity. Biomed Pharmacother 2025; 185:117944. [PMID: 40056826 DOI: 10.1016/j.biopha.2025.117944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/19/2025] [Accepted: 02/24/2025] [Indexed: 03/10/2025] Open
Abstract
PURPOSE To analyze the response of different retinal ganglion cell (RGC) populations to NMDA-induced retinal excitotoxicity and the effect of an intraperitoneal treatment with 7,8-Dihydroxyflavone (DHF), a potent selective TrkB agonist. METHODS Adult albino rats were treated the day prior to NMDA injection and the three following days with intraperitoneal vehicle (1 %DMSO in 0.09 %NaCl) or DHF (5 mg/kg in vehicle) injections. DHF-afforded protection was studied in the population of Brn3a+RGCs, OPN+RGCs (α-RGCs), OPN+ Tbr2+RGCs (αONs-RGCs), OPN+ Tbr2-Brn3a-RGCs (αONt-RGCs) and OPN+Brn3a+RGCs (αOFF-RGCs) at 3,7,14, or 21 days. The functional response was analyzed longitudinally with full-field electroretinograms. The mechanisms underlying DHF-afforded neuroprotection were assessed by western blot (WB) analysis of the levels of phosphorylated and total TrkB, phosphatidylinositol 3 kinase (PIK3/AKT) and mitogen-activated protein kinase (MAPK). RESULTS NMDA intravitreal injection resulted in a significant diminution of the mean amplitudes of the pSTR and b-waves, as well as in severe depletion of all RGCs studied except αONt-RGCs. DHF treatment resulted in rescued mean amplitudes of the pSTR and b-waves up to 21 days after NMDA. WB analysis revealed an increase in p-TrkB which correlates to the increase of TRKB protein and an increase in normalized pAKT/AKT. pMAPK/MAPK was upregulated earlier and significantly higher in DHF-treated retinas. DHF afforded survival of up to 49 % of the Brn3a+RGCs versus 25 % of the vehicle group at 21 days after NMDA, and improved survival of the α-RGC and αONs-RGCs but did not rescue the αOFF-RGCs. CONCLUSION Different RGC types exhibit variable susceptibilities to NMDA injury, and DHF-mediated activation of TrkB affords neuroprotection.
Collapse
Affiliation(s)
- Alejandro Gallego-Ortega
- Department of Ophthalmology, Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca (IMIB-Arrixaca), University of Murcia, Murcia, Spain; Stein Eye Institute, Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, USA.
| | - Caridad Galindo-Romero
- Department of Ophthalmology, Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca (IMIB-Arrixaca), University of Murcia, Murcia, Spain.
| | - Beatriz Vidal-Villegas
- Department of Ophthalmology, Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca (IMIB-Arrixaca), University of Murcia, Murcia, Spain; Moorfields Eye Hospital, London, United Kingdom
| | - José Manuel Bernal-Garro
- Department of Ophthalmology, Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca (IMIB-Arrixaca), University of Murcia, Murcia, Spain
| | - Pedro de la Villa
- Department of Systems Biology, Laboratory of Visual Neurophysiology, University of Alcalá, Alcalá de Henares, Madrid, Spain
| | - Marcelino Avilés-Trigueros
- Department of Ophthalmology, Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca (IMIB-Arrixaca), University of Murcia, Murcia, Spain
| | - Manuel Vidal-Sanz
- Department of Ophthalmology, Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca (IMIB-Arrixaca), University of Murcia, Murcia, Spain
| |
Collapse
|
2
|
Wall RV, Basavarajappa D, Klistoner A, Graham S, You Y. Mechanisms of Transsynaptic Degeneration in the Aging Brain. Aging Dis 2024; 15:2149-2167. [PMID: 39191395 PMCID: PMC11346400 DOI: 10.14336/ad.2024.03019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/19/2024] [Indexed: 08/29/2024] Open
Abstract
A prominent feature in many neurodegenerative diseases involves the spread of the pathology from the initial site of damage to anatomically and functionally connected regions of the central nervous system (CNS), referred to as transsynaptic degeneration (TSD). This review covers the possible mechanisms of both retrograde and anterograde TSD in various age-related neurodegenerative diseases, including synaptically and glial mediated changes contributing to TDS and their potential as therapeutic targets. This phenomenon is well documented in clinical and experimental studies spanning various neurodegenerative diseases and their respective models, with a significant emphasis on the visual pathway, to be explored herein. With the increase in the aging population and subsequent rise in age-related neurodegenerative diseases, it is crucial to understand the underlying mechanisms of.
Collapse
Affiliation(s)
- Roshana Vander Wall
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, 2109, Australia
| | - Devaraj Basavarajappa
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, 2109, Australia
| | - Alexander Klistoner
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, 2109, Australia
| | - Stuart Graham
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, 2109, Australia
- Save Sight Institute, Sydney University, Sydney, NSW, 2000, Australia
| | - Yuyi You
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, 2109, Australia
- Save Sight Institute, Sydney University, Sydney, NSW, 2000, Australia
| |
Collapse
|
3
|
Mesoscopic Mapping of Visual Pathway in a Female 5XFAD Mouse Model of Alzheimer's Disease. Cells 2022; 11:cells11233901. [PMID: 36497159 PMCID: PMC9740259 DOI: 10.3390/cells11233901] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/25/2022] [Accepted: 12/01/2022] [Indexed: 12/11/2022] Open
Abstract
Amyloid-β (Aβ) deposition and Aβ-induced neurodegeneration appear in the retina and retinorecipient areas in the early stages of Alzheimer's disease (AD). Although these Aβ-related changes in the retina cause damage to the visual functions, no studies have yet revealed the alterations in the visual pathways of AD. Therefore, we investigated the alterations of visual circuits in the AD mouse model using anterograde tracer cholera toxin β subunits (CTβ). Moreover, we investigated the Aβ accumulation in the retina and retinorecipient areas and the neuronal loss, and synaptic degeneration in retinorecipient areas by immunofluorescent staining of 4- and 12-month-old female 5XFAD transgenic mice. Our results demonstrated that Aβ accumulation and neurodegeneration occurred in the retina and retinorecipient regions of early and late stages of the 5XFAD mice. Retinal efferents to the suprachiasmatic nucleus and lateral geniculate nucleus were impaired in the early stage of AD. Moreover, retinal connections to the dorsal lateral geniculate nucleus and superior colliculus were degenerated in the late-stage of AD. These findings reveal the Aβ-related pathology induced visual circuit disturbances at the mesoscale level in both the early and late stages of AD and provide anatomical and functional insights into the visual circuitry of AD.
Collapse
|
4
|
Galindo-Romero C, Norte-Muñoz M, Gallego-Ortega A, Rodríguez-Ramírez KT, Lucas-Ruiz F, González-Riquelme MJ, Vidal-Sanz M, Agudo-Barriuso M. The retina of the lab rat: focus on retinal ganglion cells and photoreceptors. Front Neuroanat 2022; 16:994890. [PMID: 36213609 PMCID: PMC9538360 DOI: 10.3389/fnana.2022.994890] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 09/02/2022] [Indexed: 11/21/2022] Open
Abstract
Albino and pigmented rat strains are widely used in models to study retinal degeneration and to test new therapies. Here, we have summarized the main topographical and functional characteristics of the rat retina focussing on photoreceptors and retinal ganglion cells (RGCs), the beginning and end of the retinal circuitry, respectively. These neurons are very sensitive to injury and disease, and thus knowing their normal number, topography, and function is essential to accurately investigate on neuronal survival and protection.
Collapse
|
5
|
7,8-Dihydroxiflavone Maintains Retinal Functionality and Protects Various Types of RGCs in Adult Rats with Optic Nerve Transection. Int J Mol Sci 2021; 22:ijms222111815. [PMID: 34769247 PMCID: PMC8584116 DOI: 10.3390/ijms222111815] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 12/13/2022] Open
Abstract
To analyze the neuroprotective effects of 7,8-Dihydroxyflavone (DHF) in vivo and ex vivo, adult albino Sprague-Dawley rats were given a left intraorbital optic nerve transection (IONT) and were divided in two groups: One was treated daily with intraperitoneal (ip) DHF (5 mg/kg) (n = 24) and the other (n = 18) received ip vehicle (1% DMSO in 0.9% NaCl) from one day before IONT until processing. At 5, 7, 10, 12, 14, and 21 days (d) after IONT, full field electroretinograms (ERG) were recorded from both experimental and one additional naïve-control group (n = 6). Treated rats were analyzed 7 (n = 14), 14 (n = 14) or 21 d (n = 14) after IONT, and the retinas immune stained against Brn3a, Osteopontin (OPN) and the T-box transcription factor T-brain 2 (Tbr2) to identify surviving retinal ganglion cells (RGCs) (Brn3a+), α-like (OPN+), α-OFF like (OPN+Brn3a+) or M4-like/α-ON sustained RGCs (OPN+Tbr+). Naïve and right treated retinas showed normal ERG recordings. Left vehicle-treated retinas showed decreased amplitudes of the scotopic threshold response (pSTR) (as early as 5 d), the rod b-wave, the mixed response and the cone response (as early as 10 d), which did not recover with time. In these retinas, by day 7 the total numbers of Brn3a+RGCs, OPN+RGCs and OPN+Tbr2+RGCs decreased to less than one half and OPN+Brn3a+RGCs decreased to approximately 0.5%, and Brn3a+RGCs showed a progressive loss with time, while OPN+RGCs and OPN+Tbr2+RGCs did not diminish after seven days. Compared to vehicle-treated, the left DHF-treated retinas showed significantly greater amplitudes of the pSTR, normal b-wave values and significantly greater numbers of OPN+RGCs and OPN+Tbr2+RGCs for up to 14 d and of Brn3a+RGCs for up to 21 days. DHF affords significant rescue of Brn3a+RGCs, OPN+RGCs and OPN+Tbr2+RGCs, but not OPN+Brn3a+RGCs, and preserves functional ERG responses after IONT.
Collapse
|
6
|
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
|
7
|
Cai J, Li L, Song Y, Xu L, Mao Y, Wang H. Neuroprotective Effect of Brimonidine against Facial Nerve Crush Injury in Rats via Suppressing GFAP/PAF Activation and Neuroinflammation. ORL J Otorhinolaryngol Relat Spec 2021; 83:449-456. [PMID: 33965946 DOI: 10.1159/000514994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 02/02/2021] [Indexed: 11/19/2022]
Abstract
OBJECTIVE This study aimed to investigate the potential neuroprotective action of brimonidine against facial nerve crush injury in rats and the possible underlying mechanisms. METHODS Sixty Wistar adult rats were randomly and equally divided into 3 groups: 40 rats underwent unilateral facial nerve crush injury and were administered with either saline (intraperitoneal, n = 20) or brimonidine 1 mg/kg/day (intraperitoneal, n = 20) for 5 consecutive days. Functional and electromyographic recovery was recorded postoperatively. The facial nucleus of 5 mice in each group was analyzed for mRNA expression levels of GFAP, PAF, NT-4, P75NTR, NF-κB, TNF-α, IL-6, and α2-ARs by qRT-PCR. RESULTS Brimonidine promoted the recovery of vibrissae movement, eyelid closure, and electrophysiological function in a rat model of nerve crush injury. Hematoxylin and eosin staining and electron microscopy showed significant recovery of Schwann cells and axons in the brimonidine group. Brimonidine attenuated the crush-induced upregulation in GFAP and PAF mRNA (p < 0.05), as well as enhanced the mRNA levels of NT-4 and P75NTR (p < 0.05), while decreased the expression of NF-κB, TNF-α and IL-6 (p < 0.05). CONCLUSIONS Brimonidine could promote the recovery of facial nerve crush injury in rats via suppressing of GFAP/PAF activation and neuroinflammation and increasing neurotrophic factors. Brimonidine may be apromising candidate agent for the treatment of facial nerve injury.
Collapse
Affiliation(s)
- Jing Cai
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Liheng Li
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Otolaryngology, The First Affiliated Hospital of Huzhou University, Zhejiang, China
| | - Yongdong Song
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lei Xu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yanyan Mao
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Haibo Wang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
8
|
Pang IH, Clark AF. Inducible rodent models of glaucoma. Prog Retin Eye Res 2020; 75:100799. [PMID: 31557521 PMCID: PMC7085984 DOI: 10.1016/j.preteyeres.2019.100799] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 09/16/2019] [Accepted: 09/18/2019] [Indexed: 11/23/2022]
Abstract
Glaucoma is one of the leading causes of vision impairment worldwide. In order to further understand the molecular pathobiology of this disease and to develop better therapies, clinically relevant animal models are necessary. In recent years, both the rat and mouse have become popular models in glaucoma research. Key reasons are: many important biological similarities shared among rodent eyes and the human eye; development of improved methods to induce glaucoma and to evaluate glaucomatous damage; availability of genetic tools in the mouse; as well as the relatively low cost of rodent studies. Commonly studied rat and mouse glaucoma models include intraocular pressure (IOP)-dependent and pressure-independent models. The pressure-dependent models address the most important risk factor of elevated IOP, whereas the pressure-independent models assess "normal tension" glaucoma and other "non-IOP" related factors associated with glaucomatous damage. The current article provides descriptions of these models, their characterizations, specific techniques to induce glaucoma, mechanisms of injury, advantages, and limitations.
Collapse
Affiliation(s)
- Iok-Hou Pang
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Abbot F Clark
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, USA; Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, USA.
| |
Collapse
|
9
|
Valiente-Soriano FJ, Ortín-Martínez A, Di Pierdomenico J, García-Ayuso D, Gallego-Ortega A, Miralles de Imperial-Ollero JA, Jiménez-López M, Villegas-Pérez MP, Wheeler LA, Vidal-Sanz M. Topical Brimonidine or Intravitreal BDNF, CNTF, or bFGF Protect Cones Against Phototoxicity. Transl Vis Sci Technol 2019; 8:36. [PMID: 31890348 PMCID: PMC6919195 DOI: 10.1167/tvst.8.6.36] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 10/07/2019] [Indexed: 01/20/2023] Open
Abstract
Purpose To develop a focal photoreceptor degeneration model by blue light-emitting diode (LED)-induced phototoxicity (LIP) and investigate the protective effects of topical brimonidine (BMD) or intravitreal brain-derived neurotrophic factor (BDNF), ciliary neurotrophic factor (CNTF), or basic fibroblast growth factor (bFGF). Methods In anesthetized, dark-adapted, adult female Swiss mice, the left eye was dilated and exposed to blue light (10 seconds, 200 lux). After LIP, full-field electroretinograms (ERG) and spectral-domain optical coherence tomography (SD-OCT) were obtained longitudinally, and reactive-Iba-1+monocytic cells, TUNEL+ cells and S-opsin+ cone outer segments were examined up to 7 days. Left eyes were treated topically with BMD (1%) or vehicle, before or right after LIP, or intravitreally with BDNF (2.5 μg), CNTF (0.2 μg), bFGF (0.5 μg), or corresponding vehicle right after LIP. At 7 days, S-opsin+ cone outer segments were counted within predetermined fixed-size areas (PFA) centered on the lesion in both flattened retinas. Results SD-OCT showed a circular region in the superior-temporal left retina with progressive thinning (207.9 ± 5.6 μm to 160.7 ± 6.8 μm [7 days], n = 8), increasing TUNEL+ cells (peak at 3 days), decreasing S-opsin+ cone outer segments, and strong microglia activation. ERGs were normal by 3 days. Total S-opsin+ cones in the PFA for LIP-treated and fellow-retinas were 2330 ± 262 and 5601 ± 583 (n = 8), respectively. All neuroprotectants (n = 7–11), including topical BMD pre- or post-LIP, or intravitreal BDNF, CNTF, and bFGF, showed significantly greater S-opsin+ cone survival than their corresponding vehicle-treated groups. Conclusions LIP is a reliable, quantifiable focal photoreceptor degeneration model. Topical BMD or intravitreal BDNF, CNTF, or bFGF protect against LIP-induced cone-photoreceptor loss. Translational Relevance Topical BMD or intravitreal BDNF, CNTF, or bFGF protect cones against phototoxicity.
Collapse
Affiliation(s)
- Francisco J Valiente-Soriano
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, e Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - Arturo Ortín-Martínez
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, e Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain.,Present Address: Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Johnny Di Pierdomenico
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, e Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - Diego García-Ayuso
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, e Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - Alejandro Gallego-Ortega
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, e Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - Juan A Miralles de Imperial-Ollero
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, e Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - Manuel Jiménez-López
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, e Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - María Paz Villegas-Pérez
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, e Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - Larry A Wheeler
- Ophthalmology and Visual Science, University of Utah Health Sciences Center, Salt Lake City, UT, USA
| | - Manuel Vidal-Sanz
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, e Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| |
Collapse
|
10
|
Shabanzadeh AP, D'Onofrio PM, Magharious M, Choi KAB, Monnier PP, Koeberle PD. Modifying PTEN recruitment promotes neuron survival, regeneration, and functional recovery after CNS injury. Cell Death Dis 2019; 10:567. [PMID: 31358730 PMCID: PMC6662832 DOI: 10.1038/s41419-019-1802-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 06/07/2019] [Accepted: 06/28/2019] [Indexed: 12/13/2022]
Abstract
Phosphatase and tensin homolog (PTEN) regulates apoptosis and axonal growth in the developing and adult central nervous system (CNS). Here, we show that human PTEN C-terminal PDZ interactions play a critical role in neuronal apoptosis and axon regeneration after traumatic CNS injury and stroke, highlighted by the findings that antagonizing the PDZ-motif interactions of PTEN has therapeutic applicability for these indications. Interestingly, the death-inducing function of PTEN following ischemic insult depends on a PDZ-domain interaction with MAGI-2 and MAST205, PDZ proteins that are known to recruit PTEN to the plasma membrane and stabilize its interaction with PIP3. Treatments with a human peptide that prevents PTEN association with MAGI-2 or MAST205 increased neuronal survival in multiple stroke models, in vitro. A pro-survival effect was also observed in models of retinal ischemia, optic nerve transection, and after middle cerebral artery occlusion (MCAO) in adult rats. The human PTEN peptide also improved axonal regeneration in the crushed optic nerve. Furthermore, human PTEN peptide therapy promoted functional improvement after MCAO or retinal ischemia induced via ophthalmic artery ligation. These findings show that the human peptide-based targeting of C-terminal PTEN PDZ interactions has therapeutic potential for insults of the CNS, including trauma and stroke.
Collapse
Affiliation(s)
- Alireza Pirsaraei Shabanzadeh
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Krembil Research Institute, University Health Network, Toronto, ON, M5T 2S8, Canada
| | - Philippe Matteo D'Onofrio
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Rehabilitation Science Institute, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Mark Magharious
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Rehabilitation Science Institute, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Kyung An Brian Choi
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Philippe Patrick Monnier
- Departments of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Krembil Research Institute, University Health Network, Toronto, ON, M5T 2S8, Canada
| | - Paulo Dieter Koeberle
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, ON, M5S 1A8, Canada. .,Rehabilitation Science Institute, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
11
|
Crish SD, Schofield BR. Anterograde Tract Tracing for Assaying Axonopathy and Transport Deficits in Glaucoma. Methods Mol Biol 2018; 1695:171-185. [PMID: 29190027 DOI: 10.1007/978-1-4939-7407-8_15] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Whether to stage degeneration or investigate early pathology in glaucoma, examination of axonal structure and function is essential. There are a wide variety of methods available to investigators using animal models of glaucoma, with varying utilities depending on the questions asked. Here, we describe the use of anterograde neuronal tract tracing using cholera toxin B (CTB) for the determination of axon transport integrity of the retinofugal projection. This method reveals the structure of the retinal axons as well as the functional integrity of anterograde transport systems.
Collapse
Affiliation(s)
- Samuel D Crish
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, RGE-135, 4209 St. Rt. 44, Rootstown, OH, 44272, USA.
| | - Brett R Schofield
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, 44272, USA
| |
Collapse
|
12
|
Chiasseu M, Alarcon-Martinez L, Belforte N, Quintero H, Dotigny F, Destroismaisons L, Vande Velde C, Panayi F, Louis C, Di Polo A. Tau accumulation in the retina promotes early neuronal dysfunction and precedes brain pathology in a mouse model of Alzheimer's disease. Mol Neurodegener 2017; 12:58. [PMID: 28774322 PMCID: PMC5543446 DOI: 10.1186/s13024-017-0199-3] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 07/31/2017] [Indexed: 12/14/2022] Open
Abstract
Background Tau is an axon-enriched protein that binds to and stabilizes microtubules, and hence plays a crucial role in neuronal function. In Alzheimer’s disease (AD), pathological tau accumulation correlates with cognitive decline. Substantial visual deficits are found in individuals affected by AD including a preferential loss of retinal ganglion cells (RGCs), the neurons that convey visual information from the retina to the brain. At present, however, the mechanisms that underlie vision changes in these patients are poorly understood. Here, we asked whether tau plays a role in early retinal pathology and neuronal dysfunction in AD. Methods Alterations in tau protein and gene expression, phosphorylation, and localization were investigated by western blots, qPCR, and immunohistochemistry in the retina and visual pathways of triple transgenic mice (3xTg) harboring mutations in the genes encoding presenilin 1 (PS1M146 V), amyloid precursor protein (APPSwe), and tau (MAPTP301L). Anterograde axonal transport was assessed by intraocular injection of the cholera toxin beta subunit followed by quantification of tracer accumulation in the contralateral superior colliculus. RGC survival was analyzed on whole-mounted retinas using cell-specific markers. Reduction of tau expression was achieved following intravitreal injection of targeted siRNA. Results Our data demonstrate an age-related increase in endogenous retinal tau characterized by epitope-specific hypo- and hyper-phosphorylation in 3xTg mice. Retinal tau accumulation was observed as early as three months of age, prior to the reported onset of behavioral deficits, and preceded tau aggregation in the brain. Intriguingly, tau build up occurred in RGC soma and dendrites, while tau in RGC axons in the optic nerve was depleted. Tau phosphorylation changes and missorting correlated with substantial defects in anterograde axonal transport that preceded RGC death. Importantly, targeted siRNA-mediated knockdown of endogenous tau improved anterograde transport along RGC axons. Conclusions Our study reveals profound tau pathology in the visual system leading to early retinal neuron damage in a mouse model of AD. Importantly, we show that tau accumulation promotes anterograde axonal transport impairment in vivo, and identify this response as an early feature of neuronal dysfunction that precedes cell death in the AD retina. These findings provide the first proof-of-concept that a global strategy to reduce tau accumulation is beneficial to improve axonal transport and mitigate functional deficits in AD and tauopathies.
Collapse
Affiliation(s)
- Marius Chiasseu
- Department of Neuroscience and Centre de recherche du centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, 900 Rue Saint-Denis, Tour Viger, Room R09.720, Montréal, QC, H2X 0A9, Canada
| | - Luis Alarcon-Martinez
- Department of Neuroscience and Centre de recherche du centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, 900 Rue Saint-Denis, Tour Viger, Room R09.720, Montréal, QC, H2X 0A9, Canada
| | - Nicolas Belforte
- Department of Neuroscience and Centre de recherche du centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, 900 Rue Saint-Denis, Tour Viger, Room R09.720, Montréal, QC, H2X 0A9, Canada
| | - Heberto Quintero
- Department of Neuroscience and Centre de recherche du centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, 900 Rue Saint-Denis, Tour Viger, Room R09.720, Montréal, QC, H2X 0A9, Canada
| | - Florence Dotigny
- Department of Neuroscience and Centre de recherche du centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, 900 Rue Saint-Denis, Tour Viger, Room R09.720, Montréal, QC, H2X 0A9, Canada
| | - Laurie Destroismaisons
- Department of Neuroscience and Centre de recherche du centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, 900 Rue Saint-Denis, Tour Viger, Room R09.720, Montréal, QC, H2X 0A9, Canada
| | - Christine Vande Velde
- Department of Neuroscience and Centre de recherche du centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, 900 Rue Saint-Denis, Tour Viger, Room R09.720, Montréal, QC, H2X 0A9, Canada
| | - Fany Panayi
- Institut de Recherches Servier, 78290, Croissy-sur-Seine, France
| | - Caroline Louis
- Institut de Recherches Servier, 78290, Croissy-sur-Seine, France
| | - Adriana Di Polo
- Department of Neuroscience and Centre de recherche du centre hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, 900 Rue Saint-Denis, Tour Viger, Room R09.720, Montréal, QC, H2X 0A9, Canada.
| |
Collapse
|
13
|
Galindo-Romero C, Harun-Or-Rashid M, Jiménez-López M, Vidal-Sanz M, Agudo-Barriuso M, Hallböök F. Neuroprotection by α2-Adrenergic Receptor Stimulation after Excitotoxic Retinal Injury: A Study of the Total Population of Retinal Ganglion Cells and Their Distribution in the Chicken Retina. PLoS One 2016; 11:e0161862. [PMID: 27611432 PMCID: PMC5017579 DOI: 10.1371/journal.pone.0161862] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 07/14/2016] [Indexed: 11/28/2022] Open
Abstract
We have studied the effect of α2-adrenergic receptor stimulation on the total excitotoxically injured chicken retinal ganglion cell population. N-methyl-D-aspartate (NMDA) was intraocularly injected at embryonic day 18 and Brn3a positive retinal ganglion cells (Brn3a+ RGCs) were counted in flat-mounted retinas using automated routines. The number and distribution of the Brn3a+ RGCs were analyzed in series of normal retinas from embryonic day 8 to post-hatch day 11 retinas and in retinas 7 or 14 days post NMDA lesion. The total number of Brn3a+ RGCs in the post-hatch retina was approximately 1.9x106 with a density of approximately 9.2x103 cells/mm2. The isodensity maps of normal retina showed that the density decreased with age as the retinal size increased. In contrast to previous studies, we did not find any specific region with increased RGC density, rather the Brn3a+ RGCs were homogeneously distributed over the central retina with decreasing density in the periphery and in the region of the pecten oculli. Injection of 5–10 μg NMDA caused 30–50% loss of Brn3a+ cells and the loss was more severe in the dorsal than in the ventral retina. Pretreatment with brimonidine reduced the loss of Brn3a+ cells both 7 and 14 days post lesion and the protective effect was higher in the dorsal than in the ventral retina. We conclude that α2-adrenergic receptor stimulation reduced the impact of the excitotoxic injury in chicken similarly to what has been shown in mammals. Furthermore, the data show that the RGCs are evenly distributed over in the retina, which challenges previous results that indicate the presence of specific high RGC-density regions of the chicken retina.
Collapse
Affiliation(s)
- Caridad Galindo-Romero
- Department of Neuroscience, Uppsala University, Box 593, 751 24 Uppsala, Sweden
- Instituto Murciano de Investigación Biosanitaria Hospital Virgen de la Arrixaca (IMIB-Virgen de la Arrixaca) and Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, Murcia, Spain
| | | | - Manuel Jiménez-López
- Instituto Murciano de Investigación Biosanitaria Hospital Virgen de la Arrixaca (IMIB-Virgen de la Arrixaca) and Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, Murcia, Spain
| | - Manuel Vidal-Sanz
- Instituto Murciano de Investigación Biosanitaria Hospital Virgen de la Arrixaca (IMIB-Virgen de la Arrixaca) and Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, Murcia, Spain
| | - Marta Agudo-Barriuso
- Instituto Murciano de Investigación Biosanitaria Hospital Virgen de la Arrixaca (IMIB-Virgen de la Arrixaca) and Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, Murcia, Spain
| | - Finn Hallböök
- Department of Neuroscience, Uppsala University, Box 593, 751 24 Uppsala, Sweden
- * E-mail:
| |
Collapse
|
14
|
DeMar J, Sharrow K, Hill M, Berman J, Oliver T, Long J. Effects of Primary Blast Overpressure on Retina and Optic Tract in Rats. Front Neurol 2016; 7:59. [PMID: 27199884 PMCID: PMC4842954 DOI: 10.3389/fneur.2016.00059] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 04/08/2016] [Indexed: 12/19/2022] Open
Abstract
Blast has been the leading cause of injury, particularly traumatic brain injury and visual system injury, in combat operations in Iraq and Afghanistan. We determined the effect of shock tube-generated primary blast on retinal electrophysiology and on retinal and brain optic tract histopathology in a rat model. The amplitude of a- and b-waves on the electroretinogram (ERG) for both right and left eyes were measured prior to a battlefield simulation Friedlander-type blast wave and on 1, 7, and 14 days thereafter. Histopathologic findings of the right and left retina and the right and left optic tracts (2.8 mm postoptic chiasm) were evaluated 14 days after the blast. For two experiments in which the right eye was oriented to the blast, the amplitude of ERG a- and b-waves at 7 days post blast on the right side but not on the left side was diminished compared to that of sham animals (P = 0.005–0.01) Histopathologic injury scores at 14 days post blast for the right retina but not the left retina were higher than for sham animals (P = 0.01), and histopathologic injury scores at 14 days for both optic tracts were markedly higher than for shams (P < 0.0001). Exposure of one eye to a blast wave, comparable to that causing human injury, produced injury to the retina as determined by ERG and histopathology, and to both postchiasmatic optic tracts as determined by histopathology. This model may be useful for analyzing the effect of therapeutic interventions on retinal damage due to primary blast waves.
Collapse
Affiliation(s)
- James DeMar
- Blast Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research , Silver Spring, MD , USA
| | - Keith Sharrow
- Medical Countermeasures Systems, Ft. Detrick , Frederick, MD , USA
| | - Miya Hill
- Blast Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research , Silver Spring, MD , USA
| | - Jonathan Berman
- Clinical Pharmacology Department, Walter Reed Army Institute of Research , Silver Spring, MD , USA
| | - Thomas Oliver
- Clinical Research Unit, Uniformed Services University of the Health Science (USUHS) , Bethesda, MD , USA
| | - Joseph Long
- Blast Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research , Silver Spring, MD , USA
| |
Collapse
|
15
|
Kim BJ, Silverman SM, Liu Y, Wordinger RJ, Pang IH, Clark AF. In vitro and in vivo neuroprotective effects of cJun N-terminal kinase inhibitors on retinal ganglion cells. Mol Neurodegener 2016; 11:30. [PMID: 27098079 PMCID: PMC4839164 DOI: 10.1186/s13024-016-0093-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 04/08/2016] [Indexed: 01/24/2023] Open
Abstract
Background The c-Jun N-terminal kinase (JNK) signaling pathway plays an important role in neuronal pathophysiology. Using JNK inhibitors, we examined involvement of the JNK pathway in cultured rat retinal ganglion cell (RGC) death and in mouse retinal ischemia/reperfusion (I/R) injury of the visual axis. The in vitro effects of JNK inhibitors were evaluated in cultured adult rat retinal cells enriched in RGCs. Retinal I/R was induced in C57BL/6J mice through elevation of intraocular pressure to 120 mmHg for 60 min followed by reperfusion. SP600125 was administered intraperitoneally once daily for 28 days. Phosphorylation of JNK and c-Jun in the retina was examined by immunoblotting and immunohistochemistry. The thickness of retinal layers and cell numbers in the ganglion cell layer (GCL) were examined using H&E stained retinal cross sections and spectral domain optical coherence tomography (SD-OCT). Retinal function was measured by scotopic flash electroretinography (ERG). Volumetric measurement of the superior colliculus (SC) as well as VGLUT2 and PSD95 expression were studied. Results JNK inhibitors SP600125 and TAT-JNK-III, dose-dependently and significantly (p < 0.05) protected against glutamate excitotoxicity and trophic factor withdrawal induced RGC death in culture. In the I/R model, phosphorylation of JNK (pJNK) in the retina was significantly (p < 0.05) increased after injury. I/R injury significantly (p < 0.05) decreased the thickness of retinal layers, including the whole retina, inner plexiform layer, and inner nuclear layer and cell numbers in the GCL. Administration of SP600125 for 28 days protected against all these degenerative morphological changes (p < 0.05). In addition, SP600125 significantly (p < 0.05) protected against I/R-induced reduction in scotopic ERG b-wave amplitude at 3, 7, 14, 21 and 28 days after injury. SP600125 also protected against the I/R-induced losses in volume and levels of synaptic markers in the SC. Moreover, the protective effects of SP600125 in the retina and SC were also detected even with only 7 days (Days 1–7 after I/R) of SP600125 treatment. Conclusions Our results demonstrate the important role the JNK pathway plays in retinal degeneration in both in vitro and in vivo models and suggest that JNK inhibitors may be a useful therapeutic strategy for neuroprotection of RGCs in the retina. Electronic supplementary material The online version of this article (doi:10.1186/s13024-016-0093-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Byung-Jin Kim
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76109, USA.,Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA.,Present Address: Department of Ophthalmology, Johns Hopkins University School of Medicine, 400 N. Broadway, Baltimore, MD, 21231, USA
| | - Sean M Silverman
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76109, USA.,Department of Cell Biology & Immunology, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Yang Liu
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76109, USA.,Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA.,Department of Cell Biology & Immunology, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Robert J Wordinger
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76109, USA.,Department of Cell Biology & Immunology, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Iok-Hou Pang
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76109, USA.,Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Abbot F Clark
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76109, USA. .,Department of Cell Biology & Immunology, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA.
| |
Collapse
|
16
|
Vidal-Sanz M, Valiente-Soriano FJ, Ortín-Martínez A, Nadal-Nicolás FM, Jiménez-López M, Salinas-Navarro M, Alarcón-Martínez L, García-Ayuso D, Avilés-Trigueros M, Agudo-Barriuso M, Villegas-Pérez MP. Retinal neurodegeneration in experimental glaucoma. PROGRESS IN BRAIN RESEARCH 2015; 220:1-35. [PMID: 26497783 DOI: 10.1016/bs.pbr.2015.04.008] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In rats and mice, limbar tissues of the left eye were laser-photocoagulated (LP) and ocular hypertension (OHT) effects were investigated 1 week to 6 months later. To investigate the innermost layers, retinas were examined in wholemounts using tracing from the superior colliculi to identify retinal ganglion cells (RGCs) with intact retrograde axonal transport, melanopsin immunodetection to identify intrinsically photosensitive RGCs (m(+)RGC), Brn3a immunodetection to identify most RGCs but not m(+)RGCs, RECA1 immunodetection to examine the inner retinal vessels, and DAPI staining to detect all nuclei in the GC layer. The outer retinal layers (ORLs) were examined in cross sections analyzed morphometrically or in wholemounts to study S- and L-cones. Innervation of the superior colliculi was examined 10 days to 14 weeks after LP with orthogradely transported cholera toxin subunit B. By 2 weeks, OHT resulted in pie-shaped sectors devoid of FG(+)RGCs or Brn3a(+)RGCs but with large numbers of DAPI(+)nuclei. Brn3a(+)RGCs were significantly greater than FG(+)RGCs, indicating the survival of large numbers of RGCs with their axonal transport impaired. The inner retinal vasculature showed no abnormalities that could account for the sectorial loss of RGCs. m(+)RGCs decreased to approximately 50-51% in a diffuse loss across the retina. Cross sections showed focal areas of degeneration in the ORLs. RGC loss at 1m diminished to 20-25% and did not progress further with time, whereas the S- and L-cone populations diminished progressively up to 6m. The retinotectal projection was reduced by 10 days and did not progress further. LP-induced OHT results in retrograde degeneration of RGCs and m(+)RGCs, severe damage to the ORL, and loss of retinotectal terminals.
Collapse
Affiliation(s)
- Manuel Vidal-Sanz
- Departamento de Oftalmología, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain.
| | - Francisco J Valiente-Soriano
- Departamento de Oftalmología, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - Arturo Ortín-Martínez
- Departamento de Oftalmología, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - Francisco M Nadal-Nicolás
- Departamento de Oftalmología, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - Manuel Jiménez-López
- Departamento de Oftalmología, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - Manuel Salinas-Navarro
- Departamento de Oftalmología, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - Luis Alarcón-Martínez
- Departamento de Oftalmología, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - Diego García-Ayuso
- Departamento de Oftalmología, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - Marcelino Avilés-Trigueros
- Departamento de Oftalmología, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - Marta Agudo-Barriuso
- Departamento de Oftalmología, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - Maria P Villegas-Pérez
- Departamento de Oftalmología, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| |
Collapse
|
17
|
Abstract
Glaucoma is a group of progressive optic neuropathies, characterized by the degeneration of retinal ganglion cells related to the level of intraocular pressure and other factors. The exact pathogenesis of glaucoma is not known, and current therapeutic options are not sufficient to prevent or recover vision loss in glaucoma patients. Functional, repeatable, and easy-to-use animal models are therefore needed. Because of their inherent advantages, rodent animals, including mice and rats, have been widely developed as models to study various aspects of glaucoma and to evaluate possible novel therapies. However, no single model has been shown to emulate all aspects of glaucoma. In this review, we discuss currently available rodent animal models of glaucoma, their strengths and weaknesses, and the possible implications for current glaucoma research.
Collapse
Affiliation(s)
- Shida Chen
- From the Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou, China
| | | |
Collapse
|
18
|
Kitaoka Y, Kojima K, Munemasa Y, Sase K, Takagi H. Axonal protection by brimonidine with modulation of p62 expression in TNF-induced optic nerve degeneration. Graefes Arch Clin Exp Ophthalmol 2015; 253:1291-6. [PMID: 25863674 PMCID: PMC4521096 DOI: 10.1007/s00417-015-3005-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 03/25/2015] [Accepted: 03/26/2015] [Indexed: 12/11/2022] Open
Abstract
Purpose The p62, also called sequestosome 1 (SQSTM1), plays a crucial role in tumor necrosis factor (TNF)-induced optic nerve degeneration. Brimonidine has been shown to have protective effects on retinal ganglion cell bodies, although its role in their axons remains to be examined. We determined whether brimonidine modulates axonal loss induced by TNF and affects the expression of p62 in the optic nerve. Methods Experiments were performed on adult male Wistar rats that received an intravitreal injection of 10 ng TNF alone or simultaneous injection of TNF and 2, 20, or 200 pmol of brimonidine tartrate. The expression of p62 in the optic nerve was examined by immunoblot analysis. The effects of brimonidine on axons were evaluated by counting axon numbers 2 weeks after intravitreal injection. Results Intravitreal injection of brimonidine exerted substantial axonal protection against TNF-induced optic nerve degeneration. Immunoblot analysis showed that p62 was upregulated in the optic nerve after intravitreal injection of TNF, and that this increase was completely inhibited by brimonidine. Treatment with brimonidine alone also significantly decreased p62 protein levels in the optic nerve compared with the basal level. Conclusions These results suggest that the modulation of p62 levels in the optic nerve by brimonidine may be involved partly in its axonal protection.
Collapse
Affiliation(s)
- Yasushi Kitaoka
- Department of Ophthalmology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan,
| | | | | | | | | |
Collapse
|
19
|
Valiente-Soriano FJ, Salinas-Navarro M, Jiménez-López M, Alarcón-Martínez L, Ortín-Martínez A, Bernal-Garro JM, Avilés-Trigueros M, Agudo-Barriuso M, Villegas-Pérez MP, Vidal-Sanz M. Effects of ocular hypertension in the visual system of pigmented mice. PLoS One 2015; 10:e0121134. [PMID: 25811653 PMCID: PMC4374934 DOI: 10.1371/journal.pone.0121134] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 02/12/2015] [Indexed: 11/21/2022] Open
Abstract
To study the effects of ocular hypertension (OHT) on the visual system of C57BL/6 pigmented mice, the limbal and episcleral veins of the left eye were laser photocoagulated (LP). LP increased the intraocular pressure during the first five days (d), reaching basal values at 7d. To investigate the effect of OHT on the retinal ganglion cell (RGC) retrograde axonal transport, hydroxistilbamidine methanesulfonate (OHSt) was applied to both superior colliculi (SCi) and the retinas were dissected 2 or 4 weeks after LP. To determine RGC survival, these same retinas were immunoreacted against Brn3a (general RGC population) and melanopsin (intrinsically photosensitive RGCs, m+RGCs). To study whether OHT affected non-RGC neurons in the ganglion cell layer (GCL), RGCs were immunodetected with Brn3a and all GCL nuclei counterstained with DAPI in a group of animals examined 4 weeks post-LP. Innervation of the SCi was examined at 10 days, 8 or 14 weeks after LP with the orthogradely transported cholera toxin subunit-B. OHT resulted in diffuse and sectorial loss of OHSt+RGCs (50% at 2 weeks and 62% at 4 weeks) and in a comparable loss of Brn3a+RGCs at the same time intervals. m+RGCs decreased to 59% at 2 weeks and to 46% at 4 weeks, such loss was diffuse, did not parallel the sectorial loss of the general RGC population and was more severe in the superior-temporal retina. In the GCL, cell loss is selective for RGCs and does not affect other non-RGC neurons. The retinotectal innervation appeared significantly reduced at 10 days (55.7%) and did not progress further up to 14 weeks (46.6%). Thus, LP-induced OHT results in retrograde degeneration of RGCs and m+RGCs, as well as in the loss of CTB-labelled retinotectal terminals.
Collapse
Affiliation(s)
- Francisco J. Valiente-Soriano
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia. 30.100 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca) 30.100 Murcia, Spain
| | - Manuel Salinas-Navarro
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia. 30.100 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca) 30.100 Murcia, Spain
| | - Manuel Jiménez-López
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia. 30.100 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca) 30.100 Murcia, Spain
| | - Luis Alarcón-Martínez
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia. 30.100 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca) 30.100 Murcia, Spain
| | - Arturo Ortín-Martínez
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia. 30.100 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca) 30.100 Murcia, Spain
| | - José M. Bernal-Garro
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia. 30.100 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca) 30.100 Murcia, Spain
| | - Marcelino Avilés-Trigueros
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia. 30.100 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca) 30.100 Murcia, Spain
| | - Marta Agudo-Barriuso
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia. 30.100 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca) 30.100 Murcia, Spain
| | - María P. Villegas-Pérez
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia. 30.100 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca) 30.100 Murcia, Spain
| | - Manuel Vidal-Sanz
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia. 30.100 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca) 30.100 Murcia, Spain
- * E-mail:
| |
Collapse
|
20
|
Ortín-Martínez A, Salinas-Navarro M, Nadal-Nicolás FM, Jiménez-López M, Valiente-Soriano FJ, García-Ayuso D, Bernal-Garro JM, Avilés-Trigueros M, Agudo-Barriuso M, Villegas-Pérez MP, Vidal-Sanz M. Laser-induced ocular hypertension in adult rats does not affect non-RGC neurons in the ganglion cell layer but results in protracted severe loss of cone-photoreceptors. Exp Eye Res 2015; 132:17-33. [PMID: 25576772 DOI: 10.1016/j.exer.2015.01.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 11/19/2014] [Accepted: 01/07/2015] [Indexed: 01/09/2023]
Abstract
To investigate the long-term effects of laser-photocoagulation (LP)-induced ocular hypertension (OHT) in the innermost and outermost (outer-nuclear and outer segment)-retinal layers (ORL). OHT was induced in the left eye of adult rats. To investigate the ganglion cell layer (GCL) wholemounts were examined at 1, 3 or 6 months using Brn3a-immunodetection to identify retinal ganglion cells (RGCs) and DAPI-staining to detect all nuclei in this layer. To study the effects of LP on the ORL up to 6 months, retinas were: i) fresh extracted to quantify the levels of rod-, S- and L-opsin; ii) cut in cross-sections for morphometric analysis, or; iii) prepared as wholemounts to quantify and study retinal distributions of entire populations of RGCs (retrogradely labeled with fluorogold, FG), S- and L-cones (immunolabeled). OHT resulted in wedge-like sectors with their apex on the optic disc devoid of Brn3a(+)RGCs but with large numbers of DAPI(+)nuclei. The levels of all opsins diminished by 2 weeks and further decreased to 20% of basal-levels by 3 months. Cross-sections revealed focal areas of ORL degeneration. RGC survival at 15 days represented approximately 28% and did not change with time, whereas the S- and L-cone populations diminished to 65% and 80%, or to 20 and 35% at 1 or 6 months, respectively. In conclusion, LP induces in the GCL selective RGCs loss that does not progress after 1 month, and S- and L-cone loss that progresses for up to 6 months. Thus, OHT results in severe damage to both the innermost and the ORL.
Collapse
Affiliation(s)
- Arturo Ortín-Martínez
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), 30100 Murcia, Spain
| | - Manuel Salinas-Navarro
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), 30100 Murcia, Spain
| | - Francisco Manuel Nadal-Nicolás
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), 30100 Murcia, Spain
| | - Manuel Jiménez-López
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), 30100 Murcia, Spain
| | - Francisco Javier Valiente-Soriano
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), 30100 Murcia, Spain
| | - Diego García-Ayuso
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), 30100 Murcia, Spain
| | - José Manuel Bernal-Garro
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), 30100 Murcia, Spain
| | - Marcelino Avilés-Trigueros
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), 30100 Murcia, Spain
| | - Marta Agudo-Barriuso
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), 30100 Murcia, Spain
| | - María Paz Villegas-Pérez
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), 30100 Murcia, Spain
| | - Manuel Vidal-Sanz
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), 30100 Murcia, Spain.
| |
Collapse
|
21
|
Chidlow G, Wood JPM, Casson RJ. Expression of inducible heat shock proteins Hsp27 and Hsp70 in the visual pathway of rats subjected to various models of retinal ganglion cell injury. PLoS One 2014; 9:e114838. [PMID: 25535743 PMCID: PMC4275305 DOI: 10.1371/journal.pone.0114838] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2014] [Accepted: 11/14/2014] [Indexed: 01/09/2023] Open
Abstract
Inducible heat shock proteins (Hsps) are upregulated in the central nervous system in response to a wide variety of injuries. Surprisingly, however, no coherent picture has emerged regarding the magnitude, duration and cellular distribution of inducible Hsps in the visual system following injury to retinal ganglion cells (RGCs). The current study sought, therefore, to achieve the following two objectives. The first aim of this study was to systematically characterise the patterns of Hsp27 and −70 expression in the retina and optic nerve in four discrete models of retinal ganglion cell (RGC) degeneration: axonal injury (ON crush), somato-dendritic injury (NMDA-induced excitotoxicity), chronic hypoperfusion (bilateral occlusion of the carotid arteris) and experimental glaucoma. The second aim was to document Hsp27 and −70 expression in the optic tract, the subcortical retinorecipient areas of the brain, and the visual cortex during Wallerian degeneration of RGC axons. Hsp27 was robustly upregulated in the retina in each injury paradigm, with the chronic models, 2VO and experimental glaucoma, displaying a more persistent Hsp27 transcriptional response than the acute models. Hsp27 expression was always associated with astrocytes and with a subset of RGCs in each of the models excluding NMDA. Hsp27 was present within astrocytes of the optic nerve/optic tract in control rats. During Wallerian degeneration, Hsp27 was upregulated in the optic nerve/optic tract and expressed de novo by astrocytes in the lateral geniculate nucleus and the stratum opticum of the superior colliculus. Conversely, the results of our study indicate Hsp70 was minimally induced in any of the models of injury, either in the retina, or in the optic nerve/optic tract, or in the subcortical, retinorecipient areas of the brain. The findings of the present study augment our understanding of the involvement of Hsp27 and Hsp70 in the response of the visual system to RGC degeneration.
Collapse
Affiliation(s)
- Glyn Chidlow
- Ophthalmic Research Laboratories, South Australian Institute of Ophthalmology, Hanson Institute Centre for Neurological Diseases, Frome Road, Adelaide SA-5000, Australia
- Department of Ophthalmology and Visual Sciences, University of Adelaide, Frome Road, Adelaide SA-5000, Australia
- * E-mail:
| | - John P. M. Wood
- Ophthalmic Research Laboratories, South Australian Institute of Ophthalmology, Hanson Institute Centre for Neurological Diseases, Frome Road, Adelaide SA-5000, Australia
- Department of Ophthalmology and Visual Sciences, University of Adelaide, Frome Road, Adelaide SA-5000, Australia
| | - Robert J. Casson
- Ophthalmic Research Laboratories, South Australian Institute of Ophthalmology, Hanson Institute Centre for Neurological Diseases, Frome Road, Adelaide SA-5000, Australia
- Department of Ophthalmology and Visual Sciences, University of Adelaide, Frome Road, Adelaide SA-5000, Australia
| |
Collapse
|
22
|
Two methods to trace retinal ganglion cells with fluorogold: from the intact optic nerve or by stereotactic injection into the optic tract. Exp Eye Res 2014; 131:12-9. [PMID: 25482219 DOI: 10.1016/j.exer.2014.12.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 11/13/2014] [Accepted: 12/03/2014] [Indexed: 01/29/2023]
|
23
|
Ortín-Martínez A, Valiente-Soriano FJ, García-Ayuso D, Alarcón-Martínez L, Jiménez-López M, Bernal-Garro JM, Nieto-López L, Nadal-Nicolás FM, Villegas-Pérez MP, Wheeler LA, Vidal-Sanz M. A novel in vivo model of focal light emitting diode-induced cone-photoreceptor phototoxicity: neuroprotection afforded by brimonidine, BDNF, PEDF or bFGF. PLoS One 2014; 9:e113798. [PMID: 25464513 PMCID: PMC4252057 DOI: 10.1371/journal.pone.0113798] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Accepted: 10/30/2014] [Indexed: 11/19/2022] Open
Abstract
We have investigated the effects of light-emitting diode (LED)-induced phototoxicity (LIP) on cone-photoreceptors and their protection with brimonidine (BMD), brain-derived neurotrophic factor (BDNF), pigment epithelium-derived factor (PEDF), ciliary neurotrophic factor (CNTF) or basic fibroblast growth factor (bFGF). In anesthetized, dark adapted, adult albino rats a blue (400 nm) LED was placed perpendicular to the cornea (10 sec, 200 lux) and the effects were investigated using Spectral Domain Optical Coherence Tomography (SD-OCT) and/or analysing the retina in oriented cross-sections or wholemounts immune-labelled for L- and S-opsin and counterstained with the nuclear stain DAPI. The effects of topical BMD (1%) or, intravitreally injected BDNF (5 µg), PEDF (2 µg), CNTF (0.4 µg) or bFGF (1 µg) after LIP were examined on wholemounts at 7 days. SD-OCT showed damage in a circular region of the superotemporal retina, whose diameter varied from 1,842.4±84.5 µm (at 24 hours) to 1,407.7±52.8 µm (at 7 days). This region had a progressive thickness diminution from 183.4±5 µm (at 12 h) to 114.6±6 µm (at 7 d). Oriented cross-sections showed within the light-damaged region of the retina massive loss of rods and cone-photoreceptors. Wholemounts documented a circular region containing lower numbers of L- and S-cones. Within a circular area (1 mm or 1.3 mm radius, respectively) in the left and in its corresponding region of the contralateral-fellow-retina, total L- or S-cones were 7,118±842 or 661±125 for the LED exposed retinas (n = 7) and 14,040±1,860 or 2,255±193 for the fellow retinas (n = 7), respectively. BMD, BDNF, PEDF and bFGF but not CNTF showed significant neuroprotective effects on L- or S-cones. We conclude that LIP results in rod and cone-photoreceptor loss, and is a reliable, quantifiable model to study cone-photoreceptor degeneration. Intravitreal BDNF, PEDF or bFGF, or topical BMD afford significant cone neuroprotection in this model.
Collapse
Affiliation(s)
- Arturo Ortín-Martínez
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - Francisco Javier Valiente-Soriano
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - Diego García-Ayuso
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - Luis Alarcón-Martínez
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - Manuel Jiménez-López
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - José Manuel Bernal-Garro
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - Leticia Nieto-López
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - Francisco Manuel Nadal-Nicolás
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - María Paz Villegas-Pérez
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | - Larry A. Wheeler
- Zeteo Drug Discovery LLC, Irvine, California, United States of America
| | - Manuel Vidal-Sanz
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, and Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| |
Collapse
|
24
|
Dengler-Crish CM, Smith MA, Inman DM, Wilson GN, Young JW, Crish SD. Anterograde transport blockade precedes deficits in retrograde transport in the visual projection of the DBA/2J mouse model of glaucoma. Front Neurosci 2014; 8:290. [PMID: 25278826 PMCID: PMC4166356 DOI: 10.3389/fnins.2014.00290] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 08/27/2014] [Indexed: 01/30/2023] Open
Abstract
Axonal transport deficits have been reported as an early pathology in several neurodegenerative disorders, including glaucoma. However, the progression and mechanisms of these deficits are poorly understood. Previous work suggests that anterograde transport is affected earlier and to a larger degree than retrograde transport, yet this has never been examined directly in vivo. Using combined anterograde and retrograde tract tracing methods, we examined the time-course of anterograde and retrograde transport deficits in the retinofugal projection in pre-glaucomatous (3 month-old) and glaucomatous (9–13 month old) DBA/2J mice. DBA/2J-Gpnmb+ mice were used as a control strain and were shown to have similar retinal ganglion cell densities as C57BL/6J control mice—a strain commonly investigated in the field of vision research. Using cholera toxin-B injections into the eye and FluoroGold injections into the superior colliculus (SC), we were able to measure anterograde and retrograde transport in the primary visual projection. In DBA/2J, anterograde transport from the retina to SC was decreased by 69% in the 9–10 month-old age group, while retrograde transport was only reduced by 23% from levels seen in pre-glaucomatous mice. Despite this minor reduction, retrograde transport remained largely intact in these glaucomatous age groups until 13-months of age. These findings indicate that axonal transport deficits occur in semi-functional axons that are still connected to their brain targets. Structural persistence as determined by presence of estrogen-related receptor beta label in the superficial SC was maintained beyond time-points where reductions in retrograde transport occurred, also supporting that transport deficits may be due to physiological or functional abnormalities as opposed to overt structural loss.
Collapse
Affiliation(s)
- Christine M Dengler-Crish
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University Rootstown, OH, USA ; Department of Anatomy and Neurobiology, Northeast Ohio Medical University Rootstown, OH, USA
| | - Matthew A Smith
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University Rootstown, OH, USA ; Integrated Pharmaceutical Medicine Graduate Program, Northeast Ohio Medical University Rootstown, OH, USA
| | - Denise M Inman
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University Rootstown, OH, USA
| | - Gina N Wilson
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University Rootstown, OH, USA ; Biomedical Sciences Graduate Program, Kent State University Kent, OH, USA
| | - Jesse W Young
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University Rootstown, OH, USA
| | - Samuel D Crish
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University Rootstown, OH, USA
| |
Collapse
|
25
|
Dai Y, Lindsey JD, Duong-Polk KX, Chindasub P, Leung CKS, Weinreb RN. Brimonidine protects against loss of Thy-1 promoter activation following optic nerve crush. BMC Ophthalmol 2013; 13:26. [PMID: 23805828 PMCID: PMC3697983 DOI: 10.1186/1471-2415-13-26] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 06/07/2013] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND The loss of RGCs expressing Thy-1 after optic nerve injury has an initial phase of rapid decline followed by a longer phase with slower reduction rate. This study used longitudinal retinal imaging of mice expressing cyan fluorescent protein under control of the Thy-1 promoter (Thy1-CFP mice) to determine how the α2-adrenergic agonist brimonidine influences loss of Thy1 promoter activation. METHODS Baseline images of the fluorescent retinal neurons in 30 Thy1-CFP mice were obtained using a modified confocal scanning laser ophthalmoscope. Next, brimonidine (100 ug/kg, IP) was administered either one time immediately after optic nerve crush, or immediately after optic nerve crush and then every 2 days for four weeks. A control group received a single saline injection immediately after optic nerve crush. All animals were imaged weekly for four weeks after optic nerve crush. Loss of fluorescent retinal neurons within specific retinal areas was determined by counting. RESULTS At one week after optic nerve crush, the proportion of fluorescent retinal neurons retaining fluorescence was 44±7% of baseline in control mice, 51±6% after one brimonidine treatment, and 55±6% after brimonidine treatment every other day (P<0.05 for both brimonidine treatment groups compared to the control group). Subsequently, the number of fluorescent retinal neurons in the group that received one treatment differed insignificantly from the control group. In contrast, the number of fluorescent retinal neurons in the group that received repeated brimonidine treatments was greater than the control group by 28% at two weeks after crush and by 32% at three weeks after crush (P<0.05 at both time points). Rate analysis showed that brimonidine slowed the initial rate of fluorescent cell decline in the animals that received multiple treatments (P<0.05). Differences in the rate of loss among the treatment groups were insignificant after the second week. CONCLUSION Repeated brimonidine treatments protect against loss of fluorescence within fluorescent retinal neurons of Thy1-CFP mice after optic nerve crush. As most of fluorescent retinal neurons in this system are RGCs, these findings indicate that repeated brimonidine treatments may protect RGC health following optic nerve crush.
Collapse
Affiliation(s)
- Yi Dai
- Hamilton Glaucoma Center and Department of Ophthalmology, University of California San Diego, La Jolla, CA 92093, USA
- Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - James D Lindsey
- Hamilton Glaucoma Center and Department of Ophthalmology, University of California San Diego, La Jolla, CA 92093, USA
| | - Karen X Duong-Polk
- Hamilton Glaucoma Center and Department of Ophthalmology, University of California San Diego, La Jolla, CA 92093, USA
| | - Panida Chindasub
- Hamilton Glaucoma Center and Department of Ophthalmology, University of California San Diego, La Jolla, CA 92093, USA
| | | | - Robert N Weinreb
- Hamilton Glaucoma Center and Department of Ophthalmology, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
26
|
Cai J, Li J, Liu W, Han Y, Wang H. Alpha2-adrenergic receptors in spiral ganglion neurons may mediate protective effects of brimonidine and yohimbine against glutamate and hydrogen peroxide toxicity. Neuroscience 2013; 228:23-35. [DOI: 10.1016/j.neuroscience.2012.10.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 09/29/2012] [Accepted: 10/01/2012] [Indexed: 10/27/2022]
|
27
|
Liu Y, Tang L, Chen B. Effects of antioxidant gene therapy on retinal neurons and oxidative stress in a model of retinal ischemia/reperfusion. Free Radic Biol Med 2012; 52:909-15. [PMID: 22240151 DOI: 10.1016/j.freeradbiomed.2011.12.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Revised: 12/08/2011] [Accepted: 12/14/2011] [Indexed: 11/16/2022]
Abstract
Retinal ischemia/reperfusion (I/R) results in neuronal death and generation of reactive oxygen species. The aim of this study was to investigate the neuroprotective effect of manganese superoxide dismutase (SOD2) on retinal ganglion cells (RGCs) in an I/R-induced retinal injury model. One eye of each Wistar rat was pretreated with recombinant adeno-associated virus containing the SOD2 gene (AAV-SOD2) or recombinant AAV containing the GFP gene (AAV-GFP) by intravitreal injection 21 days before initiation of I/R injury. Retinal I/R injury was induced by elevating intraocular pressure for 1h, and reperfusion was established immediately afterward. The number of RGCs and the inner plexiform layer (IPL) thickness were measured by Fluorogold retrograde labeling and hematoxylin and eosin staining at 6 h, 24 h, 72 h, and 5 days after injury. Superoxide anion, the number of RGCs, IPL thickness, malondialdehyde (MDA) level, 8-hydroxy-2-deoxyguanosine (8-OHdG) level, MnSOD (manganese superoxide dismutase) activity, and nitrotyrosine level were measured by fluorescence staining, immunohistochemistry, and enzyme-linked immunosorbent analysis at 5 days after I/R injury. Severe RGC loss, reduced IPL thickness, reduced MnSOD activity, and increased superoxide ion, MDA, 8-OHdG, and nitrotyrosine production were observed after I/R injury. Administration of AAV-SOD2 significantly reduced the levels of superoxide ion, MDA, 8-OHdG, and nitrotyrosine and prevented the damage to RGCs and IPL. Delivery of the antioxidant gene inhibited I/R-induced RGC and IPL damage by reducing oxidative stress and nitrative stress, suggesting that MnSOD may be relevant for the neuroprotection of the inner retina from I/R-related diseases.
Collapse
Affiliation(s)
- Yu Liu
- Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, People's Republic of China
| | | | | |
Collapse
|
28
|
Vidal-Sanz M, Salinas-Navarro M, Nadal-Nicolás FM, Alarcón-Martínez L, Valiente-Soriano FJ, Miralles de Imperial J, Avilés-Trigueros M, Agudo-Barriuso M, Villegas-Pérez MP. Understanding glaucomatous damage: Anatomical and functional data from ocular hypertensive rodent retinas. Prog Retin Eye Res 2012; 31:1-27. [DOI: 10.1016/j.preteyeres.2011.08.001] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Revised: 08/23/2011] [Accepted: 08/24/2011] [Indexed: 12/24/2022]
|
29
|
Chen B, Tang L. Protective effects of catalase on retinal ischemia/reperfusion injury in rats. Exp Eye Res 2011; 93:599-606. [PMID: 21824472 DOI: 10.1016/j.exer.2011.07.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 07/01/2011] [Accepted: 07/20/2011] [Indexed: 10/18/2022]
Abstract
Retinal ischemia/reperfusion (I/R) injury causes profound tissue damage, especially retinal ganglion cell (RGC) death. The aims of the study were to investigate whether catalase (CAT) has a neuroprotective effect on RGC after I/R injury in rats, and to determine the possible antioxidant mechanism. Wistar female rats were randonmized into four groups: normal control group (Control group), retinal I/R with vehicle group (I/R with vehicle group), retinal I/R with AAV-CAT group (I/R with AAV-CAT group), and normal retina with AAV-CAT group (normal with AAV-CAT group). One eye of each rat was pretreated with recombinant adeno-associated virus containing catalase gene (I/R with AAV-CAT group or normal with AAV-CAT group) and recombinant adeno-associated virus containing GFP gene (I/R with vehicle group) by intravitreal injection 21 days before initiation of I/R injury. Retinal I/R injury was induced by elevating intraocular pressure to 100mmHg for 1h. The number of RGC and inner plexiform layer (IPL) thickness were measured by fluorogold retrograde labeling and hematoxylin and eosin staining at 6h, 24h, 72 h and 5d after injury. Hydrogen peroxide (H(2)O(2)), the number of RGC, IPL thickness, malondialdehyde(MDA), 8-hydroxy-2-deoxyguanosine (8-OHdG), CAT activity and nitrotyrosine were measured by fluorescence staining, immunohistochemistry and enzyme-linked immunosorbent assay analysis at 5 days after injury. Electroretinographic (ERG) evaluation was also used. Pretreatment of AAV-CAT significantly decreased the levels of H(2)O(2), MDA, 8-OHdG and nitrotyrosine, increased the catalase activity, and prevented the reduction of a- and b- waves in the I/R with AAV-CAT group compare with the I/R with vehicle group (p<0.01). Catalase attenuated the I/R-induced damage of RGC and IPL and retinal function. Therefore, catalase can protect the rat retina from I/R-induced injury by enhancing the antioxidative ability and reducing oxidative stress, which suggests that catalase may be relevant for the neuroprotection of inner retina from I/R-related diseases.
Collapse
Affiliation(s)
- Baihua Chen
- Department of Ophthalmology, Second Xiangya Hospital, Central South University, 139 Renming Middle Road, Changsha Hunan Province 410011, PR China
| | | |
Collapse
|
30
|
Galindo-Romero C, Avilés-Trigueros M, Jiménez-López M, Valiente-Soriano FJ, Salinas-Navarro M, Nadal-Nicolás F, Villegas-Pérez MP, Vidal-Sanz M, Agudo-Barriuso M. Axotomy-induced retinal ganglion cell death in adult mice: quantitative and topographic time course analyses. Exp Eye Res 2011; 92:377-87. [PMID: 21354138 DOI: 10.1016/j.exer.2011.02.008] [Citation(s) in RCA: 119] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Revised: 02/14/2011] [Accepted: 02/16/2011] [Indexed: 12/11/2022]
Abstract
The fate of retinal ganglion cells after optic nerve injury has been thoroughly described in rat, but not in mice, despite the fact that this species is amply used as a model to study different experimental paradigms that affect retinal ganglion cell population. Here we have analyzed, quantitatively and topographically, the course of mice retinal ganglion cells loss induced by intraorbital nerve transection. To do this, we have doubly identified retinal ganglion cells in all retinas by tracing them from their main retinorecipient area, the superior colliculi, and by their expression of BRN3A (product of Pou4f1 gene). In rat, this transcription factor is expressed by a majority of retinal ganglion cells; however in mice it is not known how many out of the whole population of these neurons express it. Thus, in this work we have assessed, as well, the total population of BRN3A positive retinal ganglion cells. These were automatically quantified in all whole-mounted retinas using a newly developed routine. In control retinas, traced-retinal ganglion cells were automatically quantified, using the previously reported method (Salinas-Navarro et al., 2009b). After optic nerve injury, though, traced-retinal ganglion cells had to be manually quantified by retinal sampling and their total population was afterwards inferred. In naïve whole-mounts, the mean (±standard deviation) total number of traced-retinal ganglion cells was 40,437(±3196) and of BRN3A positive ones was 34,697(±1821). Retinal ganglion cell loss was first significant for both markers 5 days post-axotomy and by day 21, the last time point analyzed, only 15% or 12% of traced or BRN3A positive retinal ganglion cells respectively, survived. Isodensity maps showed that, in control retinas, BRN3A and traced-retinal ganglion cells were distributed similarly, being densest in the dorsal retina along the naso-temporal axis. After axotomy the progressive loss of BRN3A positive retinal ganglion cells was diffuse and affected the entire retina. In conclusion, this is the first study assessing the values, in terms of total number and density, of the retinal ganglion cells surviving axotomy from 2 till 21 days post-lesion. Besides, we have demonstrated that BRN3A is expressed by 85.6% of the total retinal ganglion cell population, and because BRN3A positive retinal ganglion cells show the same spatial distribution and temporal course of degeneration than traced ones, BRN3A is a reliable marker to identify, quantify and assess, ex-vivo, retinal ganglion cell loss in this species.
Collapse
Affiliation(s)
- C Galindo-Romero
- Laboratorio de Oftalmología Experimental, Facultad de Medicina, Universidad de Murcia, 30100 Murcia, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Sánchez-Migallón MC, Nadal-Nicolás FM, Jiménez-López M, Sobrado-Calvo P, Vidal-Sanz M, Agudo-Barriuso M. Brain derived neurotrophic factor maintains Brn3a expression in axotomized rat retinal ganglion cells. Exp Eye Res 2011; 92:260-7. [PMID: 21315070 DOI: 10.1016/j.exer.2011.02.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Revised: 01/03/2011] [Accepted: 02/01/2011] [Indexed: 12/12/2022]
Abstract
The transcription factor Brn3a has been reported to be a good marker for adult rat retinal ganglion cells in control and injured retinas. However, it is still unclear if Brn3a expression declines progressively by the injury itself or otherwise its expression is maintained in retinal ganglion cells that, though being injured, are still alive, as might occur when assessing neuroprotective therapies. Therefore, we have automatically quantified the whole population of surviving Brn3a positive retinal ganglion cells in retinas subjected to intraorbital optic nerve transection and treated with either brain derived neurotrophic factor or vehicle. Brain derived neurotrophic factor is known to delay retinal ganglion cell death after axotomy. Thus, comparison of both groups would inform of the suitability of Brn3a as a retinal ganglion cell marker when testing neuroprotective molecules. As internal control, retinal ganglion cells were, as well, identified in all retinas by retrogradely tracing them with fluorogold. Our data show that at all the analyzed times post-lesion, the numbers of Brn3a positive retinal ganglion cells and of fluorogold positive retinal ganglion cells are significantly higher in the brain derived neurotrophic factor-treated retinas compared to the vehicle-treated ones. Moreover, detailed isodensity maps of the surviving Brn3a positive retinal ganglion cells show that a single injection of brain derived neurotrophic factor protects retinal ganglion cells throughout the entire retina. In conclusion, Brn3a is a reliable retinal ganglion cell marker that can be used to accurately measure the potential effect of a given neuroprotective therapy.
Collapse
Affiliation(s)
- M C Sánchez-Migallón
- Laboratorio de Oftalmología Experimental. Facultad de Medicina. Universidad de Murcia, Murcia, Spain
| | | | | | | | | | | |
Collapse
|
32
|
Lambert WS, Ruiz L, Crish SD, Wheeler LA, Calkins DJ. Brimonidine prevents axonal and somatic degeneration of retinal ganglion cell neurons. Mol Neurodegener 2011; 6:4. [PMID: 21232114 PMCID: PMC3035592 DOI: 10.1186/1750-1326-6-4] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Accepted: 01/13/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Brimonidine is a common drug for lowering ocular pressure and may directly protect retinal ganglion cells in glaucoma. The disease involves early loss of retinal ganglion cell transport to brain targets followed by axonal and somatic degeneration. We examined whether brimonidine preserves ganglion cell axonal transport and abates degeneration in rats with elevated ocular pressure induced by laser cauterization of the episcleral veins. RESULTS Ocular pressure was elevated unilaterally by 90% for a period of 8 weeks post- cauterization. During this time, brimonidine (1mg/kg/day) or vehicle (phosphate-buffered saline) was delivered systemically and continuously via subcutaneous pump. Animals received bilateral intravitreal injections of fluorescent cholera toxin subunit β (CTB) two days before sacrifice to assess anterograde transport. In retinas from the vehicle group, elevated pressure induced a 44% decrease in the fraction of ganglion cells with intact uptake of CTB and a 14-42% reduction in the number of immuno-labelled ganglion cell bodies, with the worst loss occurring nasally. Elevated pressure also caused a 33% loss of ganglion cell axons in vehicle optic nerves and a 70% decrease in CTB transport to the superior colliculus. Each of these components of ganglion cell degeneration was either prevented or significantly reduced in the brimonidine treatment group. CONCLUSIONS Continuous and systemic treatment with brimonidine by subcutaneous injection significantly improved retinal ganglion cell survival with exposure to elevated ocular pressure. This effect was most striking in the nasal region of the retina. Brimonidine treatment also preserved ganglion cell axon morphology, sampling density and total number in the optic nerve with elevated pressure. Consistent with improved outcome in the optic projection, brimonidine also significantly reduced the deficits in axonal transport to the superior colliculus associated with elevated ocular pressure. As transport deficits to and from retinal ganglion cell projection targets in the brain are relevant to the progression of glaucoma, the ability of brimonidine to preserve optic nerve axons and active transport suggests its neuroprotective effects are relevant not only at the cell body, but throughout the entire optic projection.
Collapse
Affiliation(s)
- Wendi S Lambert
- The Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37205, USA.
| | | | | | | | | |
Collapse
|
33
|
Cuenca N, Pinilla I, Fernández-Sánchez L, Salinas-Navarro M, Alarcón-Martínez L, Avilés-Trigueros M, de la Villa P, Miralles de Imperial J, Villegas-Pérez MP, Vidal-Sanz M. Changes in the inner and outer retinal layers after acute increase of the intraocular pressure in adult albino Swiss mice. Exp Eye Res 2010; 91:273-85. [PMID: 20650699 DOI: 10.1016/j.exer.2010.05.020] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Revised: 04/16/2010] [Accepted: 05/25/2010] [Indexed: 11/26/2022]
Abstract
In adult albino mice the effects of increased intraocular pressure on the outer retina and its circuitry was investigated at intervals ranging 3-14 weeks. Ocular hypertension (OHT) was induced by cauterizing the vessels draining the anterior part of the mice eye, as recently reported (Salinas-Navarro et al., 2009a). Electroretinographic (ERG) responses were recorded simultaneously from both eyes and compared each other prior to and at different survival intervals of 2, 8 or 12 weeks after lasering. Animals were processed at 3, 9 or 14 weeks after lasering, and radial sections were obtained in the cryostat and further processed for immunocytochemistry using antibodies against recoverin, gamma-transducin, Protein Kinase C-alpha (PKC-alpha), calbindin or synaptophysin. The synaptic ribbons were identified using an antibody against the protein bassoon, which labels photoreceptor ribbons and nuclei were identified using TO-PRO. Laser photocoagulation of the perilimbar and episcleral veins of the left eye resulted in an increase in mean intraocular pressure to approximately over twice its baseline by 24 h that was maintained for approximately five days reaching basal levels by 1 week. ERG recordings from the different groups of mice showed their a-, b-wave and scotopic threshold response (STR) amplitudes, when compared to their contralateral fellow eye, reduced to 62%, 52% and 23% at 12 weeks after lasering. Three weeks after lasering, immunostaining with recoverin and transducin antibodies could not document any changes in the outer nuclear layer (ONL) but both ON-rod bipolar and horizontal cells had lost their dendritic processes in the outer plexiform layer (OPL). Sprouting of horizontal and bipolar cell processes were observed into the ONL. Fourteen weeks after lasering, protein kinase-C antibodies showed morphologic changes of ON-rod bipolar cells and calbindin staining showed abnormal horizontal cells and a loss of their relationship with their presynaptic input. Moreover, at this time, quantitative studies indicate significant diminutions in the number of photoreceptor synaptic ribbons/100 microm, and in the thickness of the outer nuclear and plexiform layer, when compared to their fellow eyes. Increased intraocular pressure in Swiss mice results in permanent alterations of their full field ERG responses and in changes of the inner and outer retinal circuitries.
Collapse
Affiliation(s)
- Nicolás Cuenca
- Departamento de Fisiología, Genética y Microbiología, Universidad de Alicante, 03690 San Vicente del Raspeig, Alicante E-03080, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Distal axonopathy with structural persistence in glaucomatous neurodegeneration. Proc Natl Acad Sci U S A 2010; 107:5196-201. [PMID: 20194762 DOI: 10.1073/pnas.0913141107] [Citation(s) in RCA: 293] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
An early hallmark of neuronal degeneration is distal transport loss and axon pathology. Glaucoma involves the degeneration of retinal ganglion cell (RGC) neurons and their axons in the optic nerve. Here we show that, like other neurodegenerations, distal axon injury appears early in mouse glaucoma. Where RGC axons terminate in the superior colliculus, reduction of active transport follows a retinotopic pattern resembling glaucomatous vision loss. Like glaucoma, susceptibility to transport deficits increases with age and is not necessarily associated with elevated ocular pressure. Transport deficits progress distal-to-proximal, appearing in the colliculus first followed by more proximal secondary targets and then the optic tract. Transport persists through the optic nerve head before finally failing in the retina. Although axon degeneration also progresses distal-to-proximal, myelinated RGC axons and their presynaptic terminals persist in the colliculus well after transport fails. Thus, distal transport loss is predegenerative and may represent a therapeutic target.
Collapse
|
35
|
Time-course of the retinal nerve fibre layer degeneration after complete intra-orbital optic nerve transection or crush: A comparative study. Vision Res 2009; 49:2808-25. [DOI: 10.1016/j.visres.2009.08.020] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2009] [Revised: 08/19/2009] [Accepted: 08/19/2009] [Indexed: 01/25/2023]
|
36
|
A critical discussion of the rates of progression and causes of optic nerve damage in glaucoma: International Glaucoma Think Tank II: July 25-26, 2008, Florence, Italy. J Glaucoma 2009; 18:S1-21. [PMID: 19680047 DOI: 10.1097/ijg.0b013e3181aff461] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The International Glaucoma Think Tank II brought together glaucoma clinicians and researchers from all over the world to discuss current practices in glaucoma diagnosis and management, and the neurobiology of glaucoma. The meeting focused on several themes, including rates of deterioration in glaucoma patients, mechanisms of optic nerve damage, and implications for treatment. Issues such as how to measure and integrate progression information into clinical practice, screening protocols, or trials were discussed, as were promising new technologies and limitations of currently available measurement tools. Clinical applications for genetic testing were considered. Study of the neurobiology of glaucoma continues to inform our understanding of underlying degenerative processes, as well as to introduce possibilities for early detection or prevention. Many questions regarding glaucoma pathophysiology and best treatment practices remain unanswered, but with continued research and discussion, we will advance our understanding of this disease and ensure that patients receive optimal care.
Collapse
|
37
|
Ocular hypertension impairs optic nerve axonal transport leading to progressive retinal ganglion cell degeneration. Exp Eye Res 2009; 90:168-83. [PMID: 19835874 DOI: 10.1016/j.exer.2009.10.003] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2009] [Revised: 10/03/2009] [Accepted: 10/08/2009] [Indexed: 01/26/2023]
Abstract
Ocular hypertension (OHT) is the main risk factor of glaucoma, a neuropathy leading to blindness. Here we have investigated the effects of laser photocoagulation (LP)-induced OHT, on the survival and retrograde axonal transport (RAT) of adult rat retinal ganglion cells (RGC) from 1 to 12 wks. Active RAT was examined with fluorogold (FG) applied to both superior colliculi (SCi) 1 wk before processing and passive axonal diffusion with dextran tetramethylrhodamine (DTMR) applied to the optic nerve (ON) 2 d prior to sacrifice. Surviving RGCs were identified with FG applied 1 wk pre-LP or by Brn3a immunodetection. The ON and retinal nerve fiber layer were examined by RT97-neurofibrillar staining. RGCs were counted automatically and color-coded density maps were generated. OHT retinas showed absence of FG+ or DTMR+RGCs in focal, pie-shaped and diffuse regions of the retina which, by two weeks, amounted to, approximately, an 80% of RGC loss without further increase. At this time, there was a discrepancy between the total number of surviving FG-prelabelled RGCs and of DMTR+RGCs, suggesting that a large proportion of RGCs had their RAT impaired. This was further confirmed identifying surviving RGCs by their Brn3a expression. From 3 weeks onwards, there was a close correspondence of DTMR+RGCs and FG+RGCs in the same retinal regions, suggesting axonal constriction at the ON head. Neurofibrillar staining revealed, in ONs, focal degeneration of axonal bundles and, in the retinal areas lacking backlabeled RGCs, aberrant staining of RT97 characteristic of axotomy. LP-induced OHT results in a crush-like injury to ON axons leading to the anterograde and protracted retrograde degeneration of the intraocular axons and RGCs.
Collapse
|
38
|
Goldenberg-Cohen N, Dadon-Bar-El S, Hasanreisoglu M, Avraham-Lubin BCR, Dratviman-Storobinsky O, Cohen Y, Weinberger D. Possible neuroprotective effect of brimonidine in a mouse model of ischaemic optic neuropathy. Clin Exp Ophthalmol 2009; 37:718-29. [DOI: 10.1111/j.1442-9071.2009.02108.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
39
|
Parrilla-Reverter G, Agudo M, Sobrado-Calvo P, Salinas-Navarro M, Villegas-Pérez MP, Vidal-Sanz M. Effects of different neurotrophic factors on the survival of retinal ganglion cells after a complete intraorbital nerve crush injury: A quantitative in vivo study. Exp Eye Res 2009; 89:32-41. [DOI: 10.1016/j.exer.2009.02.015] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2008] [Revised: 02/10/2009] [Accepted: 02/13/2009] [Indexed: 12/12/2022]
|
40
|
Salinas-Navarro M, Jiménez-López M, Valiente-Soriano FJ, Alarcón-Martínez L, Avilés-Trigueros M, Mayor S, Holmes T, Lund RD, Villegas-Pérez MP, Vidal-Sanz M. Retinal ganglion cell population in adult albino and pigmented mice: a computerized analysis of the entire population and its spatial distribution. Vision Res 2009; 49:637-47. [PMID: 19948111 DOI: 10.1016/j.visres.2009.01.010] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2008] [Revised: 01/14/2009] [Accepted: 01/17/2009] [Indexed: 11/25/2022]
Abstract
UNLABELLED In adult Swiss albino and C57 pigmented mice, RGCs were identified with a retrogradely transported neuronal tracer applied to both optic nerves (ON) or superior colliculi (SCi). After histological processing, the retinas were prepared as whole-mounts, examined and photographed under a fluorescence microscope equipped with a motorized stage controlled by a commercial computer image analysis system: Image-Pro Plus((R)) (IPP). Retinas were imaged as a stack of 24-bit color images (140 frames per retina) using IPP with the Scope-Pro plug-in 5.0 and the images montaged to create a high-resolution composite of the retinal whole-mount when required. Single images were also processed by specific macros written in IPP that apply a sequence of filters and transformations in order to separate individual cells for automatic counting. Cell counts were later transferred to a spreadsheet for statistical analysis and used to generate a RGC density map for each retina. RESULTS The mean total numbers of RGCs labeled from the ON, in Swiss (49,493+/-3936; n=18) or C57 mice (42,658+/-1540; n=10) were slightly higher than the mean numbers of RGCs labeled from the SCi, in Swiss (48,733+/-3954; n=43) or C57 mice (41,192+/-2821; n=42), respectively. RGCs were distributed throughout the retina and density maps revealed a horizontal region in the superior retina near the optic disk with highest RGC densities. In conclusion, the population of mice RGCs may be counted automatically with a level of confidence comparable to manual counts. The distribution of RGCs adopts a form of regional specialization that resembles a horizontal visual streak.
Collapse
Affiliation(s)
- M Salinas-Navarro
- Laboratorio de Oftalmología Experimental, Facultad de Medicina, Universidad de Murcia, E-30100 Murcia, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Salinas-Navarro M, Mayor-Torroglosa S, Jiménez-López M, Avilés-Trigueros M, Holmes TM, Lund RD, Villegas-Pérez MP, Vidal-Sanz M. A computerized analysis of the entire retinal ganglion cell population and its spatial distribution in adult rats. Vision Res 2008; 49:115-26. [PMID: 18952118 DOI: 10.1016/j.visres.2008.09.029] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2008] [Revised: 09/12/2008] [Accepted: 09/16/2008] [Indexed: 10/21/2022]
Abstract
In adult albino (SD) and pigmented (PVG) rats the entire population of retinal ganglion cells (RGCs) was quantified and their spatial distribution analyzed using a computerized technique. RGCs were back-labelled from the optic nerves (ON) or the superior colliculi (SCi) with Fluorogold (FG). Numbers of RGCs labelled from the ON [SD: 82,818+/-3,949, n=27; PVG: 89,241+/-3,576, n=6) were comparable to those labelled from the SCi [SD: 81,486+/-4,340, n=37; PVG: 87,229+/-3,199; n=59]. Detailed methodology to provide cell density information at small scales demonstrated the presence of a horizontal region in the dorsal retina with highest densities, resembling a visual streak.
Collapse
Affiliation(s)
- M Salinas-Navarro
- Laboratorio de Oftalmología Experimental, Facultad de Medicina, Universidad de Murcia, E-30100 Murcia, Spain
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Tatsumi Y, Kanamori A, Nagai-Kusuhara A, Nakanishi Y, Agarwal N, Negi A, Nakamura M. Nipradilol protects rat retinal ganglion cells from apoptosis induced by serum deprivation in vitro and by diabetes in vivo. Curr Eye Res 2008; 33:683-92. [PMID: 18696344 DOI: 10.1080/02713680802323157] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
PURPOSE To investigate if nipradilol has an anti-apoptotic effect in serum-deprived RGC-5 cells and in the streptozotocin-induced diabetic rat retina. METHODS Apoptosis was quantified by activated caspase-3 immunohistochemistry or terminal dUTP nick end-labeling assay. RESULTS Nipradilol dose-dependently suppressed apoptosis in a protein kinase A- and G-dependent manner and counteracted glutamate-induced calcium entry in the RGC-5 cells and reduced apoptotic cells in the retinal ganglion cell layer of 4- and 12-week diabetic retinas compared to controls when instilled for 5 days. Removal of the nitric oxide moiety from nipradilol blocked these effects. CONCLUSIONS Nipradilol protects RGCs from apoptosis induced by serum-deprivation in vitro and by diabetes in vivo. The NO-related signaling pathway mediates the anti-apoptotic ability of nipradilol.
Collapse
Affiliation(s)
- Yasuko Tatsumi
- Department of Surgery, Division of Ophthalmology, Kobe University Graduate School of Medicine, Kobe, Japan
| | | | | | | | | | | | | |
Collapse
|
43
|
Merin S, Obolensky A, Farber MD, Chowers I. A pilot study of topical treatment with an alpha2-agonist in patients with retinal dystrophies. J Ocul Pharmacol Ther 2008; 24:80-6. [PMID: 18201138 DOI: 10.1089/jop.2007.0022] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
PURPOSE The aim of this study was to assess the neuroprotective effect of a topical alpha2-agonist in patients with retinal dystrophies. METHODS This study was a prospective, placebo-controlled, double-masked, randomized clinical trial. A total of 26 patients with retinal dystrophies were included. One (1) randomly selected eye was treated with brimonidine tartrate 0.2% twice-daily, while the fellow eye received artificial tears. Disease progression parameters tested at 6-8-month intervals throughout the study included Goldmann visual fields, contrast sensitivity, color vision, and fullfield electroretinography. RESULTS Seventeen (17) of the 26 recruited patients completed the study. Except for 1 patient with an 18-month follow-up, all patients were followed up for 24-36 months (mean, 29). At the conclusion of the study, there were no differences detected in visual acuity, color vision, and contrast sensitivity between the treated and control eyes. There was a trend, however, toward a lesser degree of visual field loss in the brimonidine-treated eyes. There was also a delay in the time required to reach a 25% visual field loss in the treated eyes. These differences were more pronounced in a subgroup of patients diagnosed as retinitis pigmentosa and with visual fields of 5 cm2 or more at baseline. CONCLUSIONS The findings of this pilot study suggest a trend for slower progression in the eyes of patients with retinal dystrophy when treated with brimonidine, according to one of the parameters that was studied (visual field loss). Further studies that include a larger number of patients and a longer follow-up period are needed to clarify and confirm the potential neuroprotective effect of alpha2-agonists in human retinal dystrophies.
Collapse
Affiliation(s)
- Saul Merin
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel.
| | | | | | | |
Collapse
|
44
|
Vidal-Sanz M, de la Villa P, Avilés-Trigueros M, Mayor-Torroglosa S, Salinas-Navarro M, Alarcón-Martínez L, Villegas-Pérez MP. Neuroprotection of retinal ganglion cell function and their central nervous system targets. Eye (Lond) 2007. [DOI: 10.1038/sj.eye.6702888] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
45
|
Hong S, Lee JE, Kim CY, Seong GJ. Agmatine protects retinal ganglion cells from hypoxia-induced apoptosis in transformed rat retinal ganglion cell line. BMC Neurosci 2007; 8:81. [PMID: 17908330 PMCID: PMC2089075 DOI: 10.1186/1471-2202-8-81] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2007] [Accepted: 10/02/2007] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Agmatine is an endogenous polyamine formed by the decarboxylation of L-arginine. We investigated the protective effects of agmatine against hypoxia-induced apoptosis of immortalized rat retinal ganglion cells (RGC-5). RGC-5 cells were cultured in a closed hypoxic chamber (5% O2) with or without agmatine. Cell viability was determined by lactate dehydrogenase (LDH) assay and apoptosis was examined by annexin V and caspase-3 assays. Expression and phosphorylation of mitogen-activated protein kinases (MAPKs; JNK, ERK p44/42, and p38) and nuclear factor-kappa B (NF-kappaB) were investigated by Western immunoblot analysis. The effects of agmatine were compared to those of brain-derived neurotrophic factor (BDNF), a well-known protective neurotrophin for retinal ganglion cells. RESULTS After 48 hours of hypoxic culture, the LDH assay showed 52.3% cell loss, which was reduced to 25.6% and 30.1% when agmatine and BDNF were administered, respectively. This observed cell loss was due to apoptotic cell death, as established by annexin V and caspase-3 assays. Although total expression of MAPKs and NF-kappaB was not influenced by hypoxic injury, phosphorylation of these two proteins was increased. Agmatine reduced phosphorylation of JNK and NF-kappaB, while BDNF suppressed phosphorylation of ERK and p38. CONCLUSION Our results show that agmatine has neuroprotective effects against hypoxia-induced retinal ganglion cell damage in RGC-5 cells and that its effects may act through the JNK and NF-kappaB signaling pathways. Our data suggest that agmatine may lead to a novel therapeutic strategy to reduce retinal ganglion cell injury related to hypoxia.
Collapse
Affiliation(s)
- Samin Hong
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Korea
| | - Jong Eun Lee
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea
| | - Chan Yun Kim
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Korea
| | - Gong Je Seong
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
46
|
Jeng YY, Lin NT, Chang PH, Huang YP, Pang VF, Liu CH, Lin CT. Retinal ischemic injury rescued by sodium 4-phenylbutyrate in a rat model. Exp Eye Res 2006; 84:486-92. [PMID: 17178414 DOI: 10.1016/j.exer.2006.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2006] [Revised: 10/24/2006] [Accepted: 11/05/2006] [Indexed: 01/26/2023]
Abstract
Retinal ischemia is a common cause of visual impairment for humans and animals. Herein, the neuroprotective effects of phenylbutyrate (PBA) upon retinal ischemic injury were investigated using a rat model. Retinal ganglion cells (RGCs) were retrograde labeled with the fluorescent tracer fluorogold (FG) applied to the superior collicoli of test Sprague-Dawley rats. High intraocular pressure and retinal ischemia were induced seven days subsequent to such FG labeling. A dose of either 100 or 400 mg/kg PBA was administered intraperitoneally to test rats at two time points, namely 30 min prior to the induction of retinal ischemia and 1 h subsequent to the cessation of the procedure inducing retinal ischemia. The test-rat retinas were collected seven days subsequent to the induction of retinal ischemia, and densities of surviving RGCs were estimated by counting FG-labeled RGCs within the retina. Histological analysis revealed that ischemic injury caused the loss of retinal RGCs and a net decrease in retinal thickness. For PBA-treated groups, almost 100% of the RGCs were preserved by a pre-ischemia treatment with PBA (at a dose of either 100 or 400 mg/kg), while post-ischemia treatment of RGCs with PBA did not lead to the preservation of RGCs from ischemic injury by PBA as determined by the counting of whole-mount retinas. Pre-ischemia treatment of RGCs with PBA (at a dose of either 100 or 400 mg/kg) significantly reduced the level of ischemia-associated loss of thickness of the total retina, especially the inner retina, and the inner plexiform layer of retina. Besides, PBA treatment significantly reduced the ischemia-induced loss of cells in the ganglion-cell layer of the retina. Taken together, these results suggest that PBA demonstrates a marked neuroprotective effect upon high intraocular pressure-induced retinal ischemia when the PBA is administered prior to ischemia induction.
Collapse
Affiliation(s)
- Yung-Yue Jeng
- Graduate Institute of Veterinary Medicine, College of Bio-resources and Agriculture, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
47
|
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
|
48
|
Danylkova NO, Alcala SR, Pomeranz HD, McLoon LK. Neuroprotective effects of brimonidine treatment in a rodent model of ischemic optic neuropathy. Exp Eye Res 2006; 84:293-301. [PMID: 17113077 DOI: 10.1016/j.exer.2006.10.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2006] [Revised: 09/05/2006] [Accepted: 10/02/2006] [Indexed: 01/26/2023]
Abstract
Ischemic optic neuropathy (ION) is a common disorder caused by disruption of the arterial blood supply to the optic nerve. It can result in significant loss of visual acuity and/or visual field. An ischemic optic nerve injury was produced in rats by intravenous injection of Rose Bengal dye followed by argon green laser application to the retinal arteries overlying the optic nerve, causing a coagulopathy within the blood vessels and disruption of optic nerve and retinal perfusion. The effect of brimonidine tartrate eye drops on survival of retinal ganglion cell axons in this experimental paradigm was studied. One eye was treated and the contralateral eye served as a control. Four groups of animals were used for this study. Group 1 received 7 days of treatment with 0.15% brimonidine tartrate eye drops twice a day prior to the ischemic injury. Group 2 animals received 0.15% brimonidine tartrate eye drops twice a day for 14 days after photocoagulation injury. Animal groups 3 and 4 received eye drops of 0.9% NaCl twice a day either daily for 7 days before injury or daily for 14 days, respectively. All rats were sacrificed 5 months after the injury to ascertain long-term optic axon survival. Coagulopathy-induced optic nerve ischemia resulted in a 71% loss of optic axons. Treatment with brimonidine daily for the 7 days prior to the injury resulted in a greater survival of optic axons, with only a 56.1% loss compared to control. Brimonidine treatment every day for 14 days after the ischemic injury did not result in a significant rescue of optic axons compared to injury alone. In summary, the application of brimonidine eye drops for one week prior to an ischemic injury resulted in a statistically significant increase in survival of optic axons within the injured optic nerves. Brimonidine treatment of the eye after the ischemic injury did not result in axon rescue, and axon loss was similar to the injured optic nerves treated with saline only. These results suggest that brimonidine may have potential use for prevention of ION in at-risk patients.
Collapse
Affiliation(s)
- Nataliya O Danylkova
- Department of Ophthalmology, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | |
Collapse
|
49
|
Resch H, Garhofer G. Topical Drug Therapy in Glaucoma. Wien Med Wochenschr 2006; 156:501-7. [PMID: 17041807 DOI: 10.1007/s10354-006-0335-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2006] [Accepted: 05/02/2006] [Indexed: 10/24/2022]
Abstract
Glaucoma is defined as a progressive optic neuropathy involving characteristic structural changes in the optic nerve head and corresponding visual field defects. Elevated intraocular pressure (IOP) is a major risk and causative factor for glaucomatous optic neuropathy. Although mechanisms other than elevated IOP may contribute to the underlying pathophysiology of glaucoma, reducing IOP remains the primary goal of therapy. Recent clinical studies have shown that decreasing the IOP can delay, or in some cases prevent progression of this chronic ocular disease. Over the past decade, several new medical therapies have become available for the treatment of glaucoma. In this article a review of topical glaucoma therapy is presented.
Collapse
Affiliation(s)
- Hemma Resch
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
50
|
Kent AR, King L, Bartholomew LR. Vitreous Concentration of Topically Applied Brimonidine-Purite 0.15%. J Ocul Pharmacol Ther 2006; 22:242-6. [PMID: 16910864 DOI: 10.1089/jop.2006.22.242] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
PURPOSE The aim of this study was to determine the vitreous brimonidine concentration of topically applied brimonidine-Purite 0.15%. METHODS Patients scheduled for a pars plana vitrectomy were invited to participate in this study after institutional review board (IRB) approval was obtained. Each patient was asked to apply brimonidine-Purite (0.15%) drops in the designated eye either every 12 h (b.i.d.; 9 patients) or every 8 h (t.i.d.; 10 patients) for the 2 weeks proceeding scheduled surgery. The importance of the last topical dose being 12 h (b.i.d. group) or 8 h (t.i.d. group) before the scheduled surgery, was emphasized. Four (4) patients served as controls and did not receive any drops. Vitreous (approximately 0.5-1.0 mL) was aspirated prior to opening the infusion line. Specimens were frozen at -68 degrees C until analyzed. RESULTS In the b.i.d. group, the mean concentration of brimonidine was 16.74 nM+/-10.33 standard deviation (range, 0.42-34.68 nM; median, 16.38); in the t.i.d. group, the mean concentration of brimonidine was 19.16 nM+/-15.40 standard deviation (range, 0.22-39.48 nM; median, 16.98). A significant difference was observed between the (no drug) control vitreous brimonidine levels and b.i.d. or t.i.d. vitreous levels (P<0.01, <0.01, respectively; n=4, 9, and 10, respectively); and in brimonidine levels between t.i.d. phakic patients and t.i.d. patients with posterior chamber lens (P=0.04; n=4 and 6, respectively). CONCLUSIONS Brimonidine-Purite 0.15%, topically applied b.i.d. or t.i.d. for 2 weeks prior to collection, acquired vitreous levels of brimonidine at or above the 2-nM concentration known to activate the neuroprotective alpha-2 receptor in animals.
Collapse
Affiliation(s)
- Alexander R Kent
- Eyecare Physicians and Surgeons, LLC, and Storm Eye Institute, Medical University of South Carolina, Charleston, SC 29403, USA.
| | | | | |
Collapse
|