1
|
Hu Y, Grodzki LM, Bartsch U. Survival and Axonal Regeneration of Retinal Ganglion Cells in a Mouse Optic Nerve Crush Model After a Cell-Based Intravitreal Co-Administration of Ciliary Neurotrophic Factor and Glial Cell Line-Derived Neurotrophic Factor at Different Post-Lesion Time Points. Cells 2025; 14:643. [PMID: 40358167 PMCID: PMC12071274 DOI: 10.3390/cells14090643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/24/2025] [Accepted: 04/26/2025] [Indexed: 05/15/2025] Open
Abstract
We recently showed, in a mouse optic nerve crush model, that a sustained cell-based intravitreal administration of ciliary neurotrophic factor (CNTF) and glial cell line-derived neurotrophic factor (GDNF) synergistically slowed the lesion-induced degeneration of retinal ganglion cells (RGCs), resulting in the presence of approximately 35% viable RGCs eight months after the lesion. However, the combinatorial neuroprotective treatment was initiated shortly after the lesion. To mimic a more clinically relevant situation, we co-administered both factors either three or five days after an intraorbital nerve crush when approximately 35% or 57% of the RGCs were degenerated, respectively. Analyses of the retinas at different time points after the lesion consistently revealed the presence of significantly more surviving RGCs in retinas co-treated with CNTF and GDNF than in retinas treated with either factor alone. For example, when the neurotrophic factors were administered five days after the nerve crush and the animals were analyzed two months after the lesion, retinas co-treated with CNTF and GDNF contained approximately 40% of the RGCs present at the start of treatment. In comparison, monotherapy with either CNTF or GDNF protected only about 15% or 10% of the RGCs present at baseline, respectively. The number of regenerating axons in the distal nerve stumps was similar in CNTF- and CNTF/GDNF-treated animals, despite the significantly higher number of rescued RGCs in the latter group. These findings have potential implications for studies aimed at developing neuroprotective treatments for optic neuropathies such as glaucoma.
Collapse
Affiliation(s)
| | | | - Udo Bartsch
- Department of Ophthalmology, Experimental Ophthalmology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (Y.H.); (L.M.G.)
| |
Collapse
|
2
|
Wang K, Liu Y, Li S, Zhao N, Qin F, Tao Y, Song Z. Unveiling the therapeutic potential and mechanisms of stanniocalcin-1 in retinal degeneration. Surv Ophthalmol 2025; 70:106-120. [PMID: 39270826 DOI: 10.1016/j.survophthal.2024.08.001] [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: 11/29/2023] [Revised: 07/30/2024] [Accepted: 08/16/2024] [Indexed: 09/15/2024]
Abstract
Retinal degeneration (RD) is a group of ocular diseases characterized by progressive photoreceptor apoptosis and visual impairment. Mitochondrial malfunction, excessive oxidative stress, and chronic activation of neuroglia collectively contribute to the development of RD. Currently, there is a lack of efficacious therapeutic interventions for RD. Stanniocalcin-1 (STC-1) is a promising candidate molecule to decelerate photoreceptor cell death. STC-1 is a secreted calcium/phosphorus regulatory protein that exerts diverse protective effects. Accumulating evidence suggests that STC-1 protects retinal cells from ischemic injury, oxidative stress, and excessive apoptosis through enhancing the expression of uncoupling protein-2 (UCP-2). Furthermore, STC-1 exerts its antiinflammatory effects by inhibiting the activation of microglia and macrophages, as well as the synthesis and secretion of proinflammatory cytokines, such as TNF-α, IL-1, and IL-6. By employing these mechanisms, STC-1 effectively shields the retinal photoreceptors and optic nerve, thereby slowing down the progression of RD. We summarize the STC-1-mediated therapeutic effects on the degenerating retina, with a particular focus on its underlying mechanisms. These findings highlight that STC-1 may act as a versatile molecule to treat degenerative retinopathy. Further research on STC-1 is imperative to establish optimal protocols for its clinical use.
Collapse
Affiliation(s)
- Kexin Wang
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China
| | - Yashuang Liu
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China
| | - Siyu Li
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China
| | - Na Zhao
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China
| | - Fangyuan Qin
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China
| | - Ye Tao
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China.
| | - Zongming Song
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China.
| |
Collapse
|
3
|
Terenzi D, Simon N, Gachomba MJM, de Peretti JL, Nazarian B, Sein J, Anton JL, Grandjean D, Baunez C, Chaminade T. Social context and drug cues modulate inhibitory control in cocaine addiction: involvement of the STN evidenced through functional MRI. Mol Psychiatry 2024; 29:3742-3751. [PMID: 38926543 PMCID: PMC11609098 DOI: 10.1038/s41380-024-02637-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 06/28/2024]
Abstract
Addictions often develop in a social context, although the influence of social factors did not receive much attention in the neuroscience of addiction. Recent animal studies suggest that peer presence can reduce cocaine intake, an influence potentially mediated, among others, by the subthalamic nucleus (STN). However, there is to date no neurobiological study investigating this mediation in humans. This study investigated the impact of social context and drug cues on brain correlates of inhibitory control in individuals with and without cocaine use disorder (CUD) using functional Magnetic Resonance Imaging (fMRI). Seventeen CUD participants and 17 healthy controls (HC) performed a novel fMRI "Social" Stop-Signal Task (SSST) in the presence or absence of an observer while being exposed to cocaine-related (vs. neutral) cues eliciting craving in drug users. The results showed that CUD participants, while slower at stopping with neutral cues, recovered control level stopping abilities with cocaine cues, while HC did not show any difference. During inhibition (Stop Correct vs Stop Incorrect), activity in the right STN, right inferior frontal gyrus (IFG), and bilateral orbitofrontal cortex (OFC) varied according to the type of cue. Notably, the presence of an observer reversed this effect in most areas for CUD participants. These findings highlight the impact of social context and drug cues on inhibitory control in CUD and the mediation of these effects by the right STN and bilateral OFC, emphasizing the importance of considering the social context in addiction research. They also comfort the STN as a potential addiction treatment target.
Collapse
Affiliation(s)
- Damiano Terenzi
- Institut de Neurosciences de la Timone, UMR 7289 CNRS & Aix-Marseille Université, Marseille, France.
| | - Nicolas Simon
- Institut de Neurosciences de la Timone, UMR 7289 CNRS & Aix-Marseille Université, Marseille, France
- SESSTIM INSERM, IRD & Aix-Marseille Université, AP-HM, Marseille, France
| | | | - Jeanne-Laure de Peretti
- Institut de Neurosciences de la Timone, UMR 7289 CNRS & Aix-Marseille Université, Marseille, France
| | - Bruno Nazarian
- Institut de Neurosciences de la Timone, UMR 7289 CNRS & Aix-Marseille Université, Marseille, France
| | - Julien Sein
- Institut de Neurosciences de la Timone, UMR 7289 CNRS & Aix-Marseille Université, Marseille, France
| | - Jean-Luc Anton
- Institut de Neurosciences de la Timone, UMR 7289 CNRS & Aix-Marseille Université, Marseille, France
| | - Didier Grandjean
- Swiss Center for Affective Science and Department of Psychology and Educational Sciences, University of Geneva, Geneva, Switzerland
| | - Christelle Baunez
- Institut de Neurosciences de la Timone, UMR 7289 CNRS & Aix-Marseille Université, Marseille, France.
| | - Thierry Chaminade
- Institut de Neurosciences de la Timone, UMR 7289 CNRS & Aix-Marseille Université, Marseille, France
| |
Collapse
|
4
|
Hu J, Zhu M, Li D, Wu Q, Le YZ. Critical Role of VEGF as a Direct Regulator of Photoreceptor Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1415:487-491. [PMID: 37440076 DOI: 10.1007/978-3-031-27681-1_71] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Vascular endothelial growth factor (VEGF or VEGF-A), a major pathogenic factor for diabetic and hypoxic blood-retina barrier (BRB) diseases, has been shown to act as a direct functional regulator for neurons in the peripheral and central nerve systems. To determine if VEGF plays a direct role in regulating retinal neuronal function, we established specific experimental procedures and examined the effect of recombinant VEGF (rVEGF) on photoreceptor function with electroretinography (ERG) in mice. In our case, rVEGF caused a significant reduction of scotopic ERG a-wave and b-wave amplitudes and photopic ERG b-wave amplitudes in a dose-dependent manner in dark-adapted wild-type (WT) mice, shortly after the intravitreal delivery of rVEGF in dark. However, the effect of rVEGF on photoreceptor function was nullified in adult Akita diabetic mice. Our data strongly suggest that VEGF is a direct regulator of photoreceptor function and VEGF upregulation contributes significantly to the diabetes-induced reduction of photoreceptor function. In this chapter, we will discuss the relevant background, key experimental procedures and results, and clinical significance of our work.
Collapse
Affiliation(s)
- Jianyan Hu
- Department of Medicine/Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Ophthalmology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Meili Zhu
- Department of Medicine/Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Dai Li
- Department of Medicine/Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- School of Optometry, Hubei University of Science and Technology, Xianning, China
| | - Qiang Wu
- Department of Ophthalmology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- Shanghai Key Laboratory of Diabetes Mellitus, Shanghai, China
| | - Yun-Zheng Le
- Department of Medicine/Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
5
|
Wang H, Peng Z, Li Y, Sahn JJ, Hodges TR, Chou TH, Liu Q, Zhou X, Jiao S, Porciatti V, Liebl DJ, Martin SF, Wen R. σ 2R/TMEM97 in retinal ganglion cell degeneration. Sci Rep 2022; 12:20753. [PMID: 36456686 PMCID: PMC9715665 DOI: 10.1038/s41598-022-24537-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 11/16/2022] [Indexed: 12/02/2022] Open
Abstract
The sigma 2 receptor (σ2R) was recently identified as an endoplasmic reticulum (ER) membrane protein known as transmembrane protein 97 (TMEM97). Studies have shown that σ2R/TMEM97 binding compounds are neuroprotective, suggesting a role of σ2R/TMEM97 in neurodegenerative processes. To understand the function of σ2R/TMEM97 in neurodegeneration pathways, we characterized ischemia-induced retinal ganglion cell (RGC) degeneration in TMEM97-/- mice and found that RGCs in TMEM97-/- mice are resistant to degeneration. In addition, intravitreal injection of a selective σ2R/TMEM97 ligand DKR-1677 significantly protects RGCs from ischemia-induced degeneration in wildtype mice. Our results provide conclusive evidence that σ2R/TMEM97 plays a role to facilitate RGC death following ischemic injury and that inhibiting the function of σ2R/TMEM97 is neuroprotective. This work is a breakthrough toward elucidating the biology and function of σ2R/TMEM97 in RGCs and likely in other σ2R/TMEM97 expressing neurons. Moreover, these findings support future studies to develop new neuroprotective approaches for RGC degenerative diseases by inhibiting σ2R/TMEM97.
Collapse
Affiliation(s)
- Hua Wang
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Zhiyou Peng
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Yiwen Li
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - James J Sahn
- Department of Chemistry and Biochemistry, University of Texas at Austin, Austin, TX, 78712, USA
| | - Timothy R Hodges
- Department of Chemistry and Biochemistry, University of Texas at Austin, Austin, TX, 78712, USA
| | - Tsung-Han Chou
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Qiong Liu
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Xuezhi Zhou
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Shuliang Jiao
- Department of Biomedical Engineering, Florida International University, Miami, FL, 33174, USA
| | - Vittorio Porciatti
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Daniel J Liebl
- Department of Neurosurgery, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Stephen F Martin
- Department of Chemistry and Biochemistry, University of Texas at Austin, Austin, TX, 78712, USA.
| | - Rong Wen
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
6
|
Ciliary neurotrophic factor-mediated neuroprotection involves enhanced glycolysis and anabolism in degenerating mouse retinas. Nat Commun 2022; 13:7037. [PMID: 36396639 PMCID: PMC9672129 DOI: 10.1038/s41467-022-34443-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 10/25/2022] [Indexed: 11/18/2022] Open
Abstract
Ciliary neurotrophic factor (CNTF) acts as a potent neuroprotective cytokine in multiple models of retinal degeneration. To understand mechanisms underlying its broad neuroprotective effects, we have investigated the influence of CNTF on metabolism in a mouse model of photoreceptor degeneration. CNTF treatment improves the morphology of photoreceptor mitochondria, but also leads to reduced oxygen consumption and suppressed respiratory chain activities. Molecular analyses show elevated glycolytic pathway gene transcripts and active enzymes. Metabolomics analyses detect significantly higher levels of ATP and the energy currency phosphocreatine, elevated glycolytic pathway metabolites, increased TCA cycle metabolites, lipid biosynthetic pathway intermediates, nucleotides, and amino acids. Moreover, CNTF treatment restores the key antioxidant glutathione to the wild type level. Therefore, CNTF significantly impacts the metabolic status of degenerating retinas by promoting aerobic glycolysis and augmenting anabolic activities. These findings reveal cellular mechanisms underlying enhanced neuronal viability and suggest potential therapies for treating retinal degeneration.
Collapse
|
7
|
Hu J, Zhu M, Li D, Wu Q, Le YZ. VEGF as a Direct Functional Regulator of Photoreceptors and Contributing Factor to Diabetes-Induced Alteration of Photoreceptor Function. Biomolecules 2021; 11:988. [PMID: 34356612 PMCID: PMC8301820 DOI: 10.3390/biom11070988] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/02/2021] [Accepted: 07/03/2021] [Indexed: 12/15/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) is a major therapeutic target for blood-retina barrier (BRB) breakdown in diabetic retinopathy (DR), age-related macular degeneration (AMD), and other hypoxic retinal vascular disorders. To determine whether VEGF is a direct regulator of retinal neuronal function and its potential role in altering vision during the progression of DR, we examined the immediate impact of recombinant VEGF (rVEGF) on photoreceptor function with electroretinography in C57BL6 background wild-type (WT) and Akita spontaneous diabetic mice. Shortly after intravitreal injections, rVEGF caused a significant reduction of scotopic ERG a-wave and b-wave amplitudes and photopic ERG b-wave amplitudes in a dose-dependent manner in dark-adapted 1.5-mo-old WT mice. Compared with WT controls, 5-mo-old Akita spontaneous diabetic mice demonstrated a significant reduction in scotopic ERG a-wave and b-wave amplitudes and photopic ERG b-wave amplitudes. However, the effect of rVEGF altered photoreceptor function in WT controls was diminished in 5-mo-old Akita spontaneous diabetic mice. In conclusion, our results suggest that VEGF is a direct functional regulator of photoreceptors and VEGF up-regulation in DR is a contributing factor to diabetes-induced alteration of photoreceptor function. This information is critical to the understanding of the therapeutic effect and to the care of anti-VEGF drug-treated patients for BRB breakdown in DR, AMD, and other hypoxic retinal vascular disorders.
Collapse
Affiliation(s)
- Jianyan Hu
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (J.H.); (M.Z.); (D.L.)
- Department of Ophthalmology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Meili Zhu
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (J.H.); (M.Z.); (D.L.)
| | - Dai Li
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (J.H.); (M.Z.); (D.L.)
- School of Optometry, Hubei University of Science and Technology, Xianning 437100, China
| | - Qiang Wu
- Department of Ophthalmology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
- Shanghai Key Laboratory of Diabetes Mellitus, Shanghai 200233, China
| | - Yun-Zheng Le
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (J.H.); (M.Z.); (D.L.)
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
8
|
Retinal Ganglion Cell Transplantation: Approaches for Overcoming Challenges to Functional Integration. Cells 2021; 10:cells10061426. [PMID: 34200991 PMCID: PMC8228580 DOI: 10.3390/cells10061426] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/01/2021] [Accepted: 06/03/2021] [Indexed: 02/07/2023] Open
Abstract
As part of the central nervous system, mammalian retinal ganglion cells (RGCs) lack significant regenerative capacity. Glaucoma causes progressive and irreversible vision loss by damaging RGCs and their axons, which compose the optic nerve. To functionally restore vision, lost RGCs must be replaced. Despite tremendous advancements in experimental models of optic neuropathy that have elucidated pathways to induce endogenous RGC neuroprotection and axon regeneration, obstacles to achieving functional visual recovery through exogenous RGC transplantation remain. Key challenges include poor graft survival, low donor neuron localization to the host retina, and inadequate dendritogenesis and synaptogenesis with afferent amacrine and bipolar cells. In this review, we summarize the current state of experimental RGC transplantation, and we propose a set of standard approaches to quantifying and reporting experimental outcomes in order to guide a collective effort to advance the field toward functional RGC replacement and optic nerve regeneration.
Collapse
|
9
|
Bucher F, Aguilar E, Marra KV, Rapp J, Arnold J, Diaz-Aguilar S, Lange C, Agostini H, Schlunck G, Stahl A, Friedlander M. CNTF Prevents Development of Outer Retinal Neovascularization Through Upregulation of CxCl10. Invest Ophthalmol Vis Sci 2021; 61:20. [PMID: 32780864 PMCID: PMC7441336 DOI: 10.1167/iovs.61.10.20] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Purpose Ciliary neurotrophic factor (CNTF) is a well-characterized neurotrophic factor currently in clinical trials for the treatment of macular telangiectasia type II. Our previous work showed that CNTF-induced STAT3 signaling is a potent inhibitor of pathologic preretinal neovascular tuft formation in the mouse model of oxygen-induced retinopathy. In this study, we investigated the effect of CNTF on outer retinal and choroidal angiogenesis and the mechanisms that underpin the observed decrease in outer retinal neovascularization following CNTF treatment. Methods In the Vldlr–/– and laser-CNV mouse models, mice received a one-time injection (on postnatal day [P] 12 in the Vldlr–/– model and 1 day after laser in the Choroidal Neovascularization (CNV) model) of recombinant CNTF or CxCl10, and the extent of neovascular lesions was assessed 6 days posttreatment. STAT3 downstream targets affected by CNTF treatment were identified using quantitative PCR analysis. A proteome array was used to compare media conditioned by CNTF-treated and control-treated primary Müller cells to screen for CNTF-induced changes in secreted angiogenic factors. Results Intravitreal treatment with recombinant CNTF led to significant reduction in neovascularization in the Vldlr–/– and laser-CNV mouse models. Treatment effect in the Vldlr–/– was long-lasting but time sensitive, requiring intravitreal treatment before P19. Mechanistic workup in vitro as well as in vivo confirmed significant activation of the STAT3-signaling pathway in Müller cells in response to CNTF treatment and upregulation of CxCl10. Intravitreal injections of recombinant CxCl10 significantly reduced outer retinal neovascularization in vivo in both the Vldlr–/– and laser-CNV mouse models. Conclusions CNTF treatment indirectly affects outer retinal and choroidal neovascularization by inducing CxCl10 secretion from retinal Müller cells.
Collapse
Affiliation(s)
- Felicitas Bucher
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States.,Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Edith Aguilar
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States
| | - Kyle V Marra
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States.,Department of Bioengineering, University of California, San Diego, San Diego, California, United States
| | - Julian Rapp
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jakob Arnold
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sophia Diaz-Aguilar
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States.,Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Clemens Lange
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hansjürgen Agostini
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Günther Schlunck
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andreas Stahl
- Department of Ophthalmology, University Medical Center Greifswald, Greifswald, Germany
| | - Martin Friedlander
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States.,The Lowy Medical Research Institute, La Jolla, California, United States
| |
Collapse
|
10
|
Potential neuroprotective biomolecules in ophthalmology. Int Ophthalmol 2020; 41:1103-1109. [PMID: 33180279 DOI: 10.1007/s10792-020-01634-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 10/29/2020] [Indexed: 10/23/2022]
Abstract
PURPOSES Retinal neurodegenerative diseases are responsible for a huge number of ocular problems worldwide. It seems that the progression of these diseases can be managed by the application of neuroprotective molecules particularly in the early stages. This article focuses on the most common neuroprotective bioagents under investigation in ophthalmology. METHODS We searched the web of science, PubMed and Scopus databases with these keywords: "glaucoma," "diabetic retinopathy," "age-related macular degeneration," "optic neuropathy and retinal degeneration" and/or "neuroprotection." RESULTS The most commonly utilized neuroprotective drugs for ophthalmology diseases were introduced in this study. It seems that these agents can be divided into three categories according to their mechanism of action: (A) neurotrophins, (B) decreasing effect on intraocular pressure and (C) inhibition of retinal neuron apoptosis. CONCLUSION A broad range of drugs has been illustrated in the literature for treatment of neuro-ophthalmic diseases. A good classification of the most applied drugs in this field can help specialists to prescribe the best matched drug considering the stage and progression of disease. However, controlled clinical trials are needed for better evaluation of the effects of these products.
Collapse
|
11
|
Itkonen J, Annala A, Tavakoli S, Arango-Gonzalez B, Ueffing M, Toropainen E, Ruponen M, Casteleijn MG, Urtti A. Characterization, Stability, and in Vivo Efficacy Studies of Recombinant Human CNTF and Its Permeation into the Neural Retina in ex Vivo Organotypic Retinal Explant Culture Models. Pharmaceutics 2020; 12:pharmaceutics12070611. [PMID: 32629980 PMCID: PMC7408322 DOI: 10.3390/pharmaceutics12070611] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 06/26/2020] [Accepted: 06/28/2020] [Indexed: 12/17/2022] Open
Abstract
Ciliary neurotrophic factor (CNTF) is one of the most studied neuroprotective agents with acknowledged potential in treating diseases of the posterior eye segment. Although its efficacy and mechanisms of action in the retina have been studied extensively, it is still not comprehensively understood which retinal cells mediate the therapeutic effects of CNTF. As with therapeutic proteins in general, it is poorly elucidated whether exogenous CNTF administered into the vitreous can enter and distribute into the retina and hence reach potentially responsive target cells. Here, we have characterized our purified recombinant human CNTF (rhCNTF), studied the protein’s in vitro bioactivity in a cell-based assay, and evaluated the thermodynamic and oligomeric status of the protein during storage. Biological activity of rhCNTF was further evaluated in vivo in an animal model of retinal degeneration. The retinal penetration and distribution of rhCNTF after 24 h was studied utilizing two ex vivo retina models. Based on our characterization findings, our rhCNTF is correctly folded and biologically active. Moreover, based on initial screening and subsequent follow-up, we identified two buffers in which rhCNTF retains its stability during storage. Whereas rhCNTF did not show photoreceptor preservative effect or improve the function of photoreceptors in vivo, this could possibly be due to the used disease model or the short duration of action with a single intravitreal injection of rhCNTF. On the other hand, the lack of in vivo efficacy was shown to not be due to distribution limitations; permeation into the retina was observed in both retinal explant models as in 24 h rhCNTF penetrated the inner limiting membrane, and being mostly observed in the ganglion cell layer, distributed to different layers of the neural retina. As rhCNTF can reach deeper retinal layers, in general, having direct effects on resident CNTF-responsive target cells is plausible.
Collapse
Affiliation(s)
- Jaakko Itkonen
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, 00790 Helsinki, Finland; (S.T.); (M.G.C.)
- Correspondence: (J.I.); (A.U.)
| | - Ada Annala
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1, 70211 Kuopio, Finland; (A.A.); (E.T.); (M.R.)
- Utrecht Institute for Pharmaceutical Science, Utrecht University, David de Wiedgebouw, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Shirin Tavakoli
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, 00790 Helsinki, Finland; (S.T.); (M.G.C.)
| | - Blanca Arango-Gonzalez
- Institute for Ophthalmic Research, Centre for Ophthalmology, University of Tübingen, Elfriede-Aulhorn-Strasse 7, D-72076 Tübingen, Germany; (B.A.-G.); (M.U.)
| | - Marius Ueffing
- Institute for Ophthalmic Research, Centre for Ophthalmology, University of Tübingen, Elfriede-Aulhorn-Strasse 7, D-72076 Tübingen, Germany; (B.A.-G.); (M.U.)
| | - Elisa Toropainen
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1, 70211 Kuopio, Finland; (A.A.); (E.T.); (M.R.)
| | - Marika Ruponen
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1, 70211 Kuopio, Finland; (A.A.); (E.T.); (M.R.)
| | - Marco G. Casteleijn
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, 00790 Helsinki, Finland; (S.T.); (M.G.C.)
- VTT Technical Research Centre of Finland Ltd., Solutions for Natural Resources and Environment, Tietotie 2, Espoo, P.O. Box 1000, FI-02044 VTT, Finland
| | - Arto Urtti
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, 00790 Helsinki, Finland; (S.T.); (M.G.C.)
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1, 70211 Kuopio, Finland; (A.A.); (E.T.); (M.R.)
- Laboratory of Biohybrid Technologies, Institute of Chemistry, St. Petersburg State University, Universitetskii pr. 26, Peterhoff, 198504 St. Petersburg, Russia
- Correspondence: (J.I.); (A.U.)
| |
Collapse
|
12
|
Devoldere J, Peynshaert K, Dewitte H, Vanhove C, De Groef L, Moons L, Özcan SY, Dalkara D, De Smedt SC, Remaut K. Non-viral delivery of chemically modified mRNA to the retina: Subretinal versus intravitreal administration. J Control Release 2019; 307:315-330. [PMID: 31265881 DOI: 10.1016/j.jconrel.2019.06.042] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 05/20/2019] [Accepted: 06/29/2019] [Indexed: 12/20/2022]
Abstract
mRNA therapeutics have recently experienced a new wave of interest, mainly due to the discovery that chemical modifications to mRNA's molecular structure could drastically reduce its inherent immunogenicity and perceived instability. On this basis, we aimed to explore the potential of chemically stabilized mRNA for ocular applications. More specifically, we investigated the behavior of mRNA-loaded lipid-based carriers in human retinal cells (in vitro), in bovine retinal explants (ex vivo) and in mouse retinas (in vivo). We demonstrate a clear superiority of mRNA over pDNA to induce protein expression in different retinal cell types, which was further enhanced by chemical modification of the mRNA, providing up to ~1800-fold higher reporter gene expression compared to pDNA. Moreover, transgene expression could be detected for at least 20 days after a single administration of chemically modified mRNA in vitro. We furthermore determined the localization and extent of mRNA expression depending on the administration route. After subretinal (SR) administration, mRNA expression was observed in vivo and ex vivo. By contrast, intravitreal (IVT) administration resulted in limited expression in vivo. Using ex vivo bovine explants with an intact vitreoretinal (VR) interface we could attribute this to the inner limiting membrane (ILM), which presents a large barrier for non-viral delivery of mRNA, trapping mRNA complexes at the vitreal side. When the vitreous was removed, which compromises the ILM, mRNA expression was apparent and seemed to colocalize with Müller cells or photoreceptors after respectively IVT or SR administration. Taken together, this study represents a first step towards mRNA-mediated therapy for retinal diseases.
Collapse
Affiliation(s)
- Joke Devoldere
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Karen Peynshaert
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Heleen Dewitte
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Medical School of the Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1050 Jette, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - Christian Vanhove
- Department of Respiratory Medicine, Ghent University, 9000 Ghent, Belgium
| | - Lies De Groef
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Leuven, Belgium
| | - Lieve Moons
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Leuven, Belgium
| | - Sinem Yilmaz Özcan
- Neurological Sciences and Psychiatry Institute; Hacettepe University, Ankara, Turkey
| | - Deniz Dalkara
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France
| | - Stefaan C De Smedt
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - Katrien Remaut
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium.
| |
Collapse
|
13
|
Devoldere J, Peynshaert K, De Smedt SC, Remaut K. Müller cells as a target for retinal therapy. Drug Discov Today 2019; 24:1483-1498. [PMID: 30731239 DOI: 10.1016/j.drudis.2019.01.023] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/20/2018] [Accepted: 01/30/2019] [Indexed: 12/28/2022]
Abstract
Müller cells are specialized glial cells that span the entire retina from the vitreous cavity to the subretinal space. Their functional diversity and unique radial morphology render them particularly interesting targets for new therapeutic approaches. In this review, we reflect on various possibilities for selective Müller cell targeting and describe how some of their cellular mechanisms can be used for retinal neuroprotection. Intriguingly, cross-species investigation of their properties has revealed that Müller cells also have an essential role in retinal regeneration. Although many questions regarding this subject remain, it is clear that Müller cells have unique characteristics that make them suitable targets for the prevention and treatment of numerous retinal diseases.
Collapse
Affiliation(s)
- Joke Devoldere
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Karen Peynshaert
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Stefaan C De Smedt
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - Katrien Remaut
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium.
| |
Collapse
|
14
|
Delplace V, Ortin-Martinez A, Tsai ELS, Amin AN, Wallace V, Shoichet MS. Controlled release strategy designed for intravitreal protein delivery to the retina. J Control Release 2019; 293:10-20. [DOI: 10.1016/j.jconrel.2018.11.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 11/05/2018] [Accepted: 11/09/2018] [Indexed: 12/25/2022]
|
15
|
Li S, Sato K, Gordon WC, Sendtner M, Bazan NG, Jin M. Ciliary neurotrophic factor (CNTF) protects retinal cone and rod photoreceptors by suppressing excessive formation of the visual pigments. J Biol Chem 2018; 293:15256-15268. [PMID: 30115683 DOI: 10.1074/jbc.ra118.004008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 08/15/2018] [Indexed: 12/25/2022] Open
Abstract
The retinal pigment epithelium (RPE)-dependent visual cycle provides 11-cis-retinal to opsins in the photoreceptor outer segments to generate functional visual pigments that initiate phototransduction in response to light stimuli. Both RPE65 isomerase of the visual cycle and the rhodopsin visual pigment have recently been identified as critical players in mediating light-induced retinal degeneration. These findings suggest that the expression and function of RPE65 and rhodopsin need to be coordinately controlled to sustain normal vision and to protect the retina from photodamage. However, the mechanism controlling the development of the retinal visual system remains poorly understood. Here, we show that deficiency in ciliary neurotrophic factor (CNTF) up-regulates the levels of rod and cone opsins accompanied by an increase in the thickness of the outer nuclear layers and the lengths of cone and rod outer segments in the mouse retina. Moreover, retinoid isomerase activity, expression levels of RPE65 and lecithin:retinol acyltransferase (LRAT), which synthesizes the RPE65 substrate, were also significantly increased in the Cntf -/- RPE. Rod a-wave and cone b-wave amplitudes of electroretinograms were increased in Cntf -/- mice, but rod b-wave amplitudes were unchanged compared with those in WT mice. Up-regulated RPE65 and LRAT levels accelerated both the visual cycle rate and recovery rate of rod light sensitivity in Cntf -/- mice. Of note, rods and cones in Cntf -/- mice exhibited hypersusceptibility to light-induced degeneration. These results indicate that CNTF is a common extracellular factor that prevents excessive production of opsins, the photoreceptor outer segments, and 11-cis-retinal to protect rods and cones from photodamage.
Collapse
Affiliation(s)
- Songhua Li
- From the Neuroscience Center of Excellence and
| | - Kota Sato
- From the Neuroscience Center of Excellence and
| | - William C Gordon
- From the Neuroscience Center of Excellence and.,Department of Ophthalmology, Louisiana State University School of Medicine, New Orleans, Louisiana 70112 and
| | - Michael Sendtner
- the Institute of Clinical Neurobiology, University Hospital Würzburg, D-97078 Würzburg, Germany
| | - Nicolas G Bazan
- From the Neuroscience Center of Excellence and.,Department of Ophthalmology, Louisiana State University School of Medicine, New Orleans, Louisiana 70112 and
| | - Minghao Jin
- From the Neuroscience Center of Excellence and .,Department of Ophthalmology, Louisiana State University School of Medicine, New Orleans, Louisiana 70112 and
| |
Collapse
|
16
|
Pardue MT, Allen RS. Neuroprotective strategies for retinal disease. Prog Retin Eye Res 2018; 65:50-76. [PMID: 29481975 PMCID: PMC6081194 DOI: 10.1016/j.preteyeres.2018.02.002] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 02/14/2018] [Accepted: 02/20/2018] [Indexed: 12/20/2022]
Abstract
Diseases that affect the eye, including photoreceptor degeneration, diabetic retinopathy, and glaucoma, affect 11.8 million people in the US, resulting in vision loss and blindness. Loss of sight affects patient quality of life and puts an economic burden both on individuals and the greater healthcare system. Despite the urgent need for treatments, few effective options currently exist in the clinic. Here, we review research on promising neuroprotective strategies that promote neuronal survival with the potential to protect against vision loss and retinal cell death. Due to the large number of neuroprotective strategies, we restricted our review to approaches that we had direct experience with in the laboratory. We focus on drugs that target survival pathways, including bile acids like UDCA and TUDCA, steroid hormones like progesterone, therapies that target retinal dopamine, and neurotrophic factors. In addition, we review rehabilitative methods that increase endogenous repair mechanisms, including exercise and electrical stimulation therapies. For each approach, we provide background on the neuroprotective strategy, including history of use in other diseases; describe potential mechanisms of action; review the body of research performed in the retina thus far, both in animals and in humans; and discuss considerations when translating each treatment to the clinic and to the retina, including which therapies show the most promise for each retinal disease. Despite the high incidence of retinal diseases and the complexity of mechanisms involved, several promising neuroprotective treatments provide hope to prevent blindness. We discuss attractive candidates here with the goal of furthering retinal research in critical areas to rapidly translate neuroprotective strategies into the clinic.
Collapse
Affiliation(s)
- Machelle T Pardue
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, 1670 Clairmont Road, Decatur, GA, 30033, USA; Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive, Atlanta, GA, 30332, USA.
| | - Rachael S Allen
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, 1670 Clairmont Road, Decatur, GA, 30033, USA
| |
Collapse
|
17
|
Progress in Gene Therapy to Prevent Retinal Ganglion Cell Loss in Glaucoma and Leber's Hereditary Optic Neuropathy. Neural Plast 2018; 2018:7108948. [PMID: 29853847 PMCID: PMC5954906 DOI: 10.1155/2018/7108948] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/04/2018] [Indexed: 12/24/2022] Open
Abstract
The eye is at the forefront of the application of gene therapy techniques to medicine. In the United States, a gene therapy treatment for Leber's congenital amaurosis, a rare inherited retinal disease, recently became the first gene therapy to be approved by the FDA for the treatment of disease caused by mutations in a specific gene. Phase III clinical trials of gene therapy for other single-gene defect diseases of the retina and optic nerve are also currently underway. However, for optic nerve diseases not caused by single-gene defects, gene therapy strategies are likely to focus on slowing or preventing neuronal death through the expression of neuroprotective agents. In addition to these strategies, there has also been recent interest in the potential use of precise genome editing techniques to treat ocular disease. This review focuses on recent developments in gene therapy techniques for the treatment of glaucoma and Leber's hereditary optic neuropathy (LHON). We discuss recent successes in clinical trials for the treatment of LHON using gene supplementation therapy, promising neuroprotective strategies that have been employed in animal models of glaucoma and the potential use of genome editing techniques in treating optic nerve disease.
Collapse
|
18
|
Photoreceptor Protection by Mesencephalic Astrocyte-Derived Neurotrophic Factor (MANF). eNeuro 2018; 5:eN-CFN-0109-18. [PMID: 29687079 PMCID: PMC5909182 DOI: 10.1523/eneuro.0109-18.2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 04/09/2018] [Indexed: 12/21/2022] Open
Abstract
Retinal degenerations are a major cause of vision impairment and blindness. Neuroprotective therapy is a promising therapeutic strategy for retinal degenerative diseases. We investigated a novel neurotrophic factor mesencephalic astrocyte-derived neurotrophic factor (MANF) in the retina. MANF is expressed at a high level during postnatal development and the expression declines to a lower level as the retina matures. Müller cells are the major cells expressing MANF. It is also found in the retinal ganglion cells, in the inner nuclear layer (INL) neurons, and in retinal pigment epithelial (RPE) cells. Intravitreal injection of recombinant human (rh)MANF significantly protected rod and cone photoreceptors in rats carrying the rhodopsin S334ter mutation, and preserved electroretinograms (ERGs) in the rd10 (Pde6brd10/rd10 ) mice. These results indicate that MANF is a native protein in the retina and is a potent neurotrophic factor for photoreceptor protection.
Collapse
|
19
|
Heo JH, Yoon JA, Ahn EK, Kim H, Urm SH, Oak CO, Yu BC, Lee SJ. Intraperitoneal administration of adipose tissue-derived stem cells for the rescue of retinal degeneration in a mouse model via indigenous CNTF up-regulation by IL-6. J Tissue Eng Regen Med 2018; 12:e1370-e1382. [PMID: 28715614 DOI: 10.1002/term.2522] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 04/05/2017] [Accepted: 06/20/2017] [Indexed: 12/21/2022]
Abstract
As the world's population begins to age, retinal degeneration is an increasing problem, and various treatment modalities are being developed. However, there have been no therapies for degenerative retinal conditions that are not characterized by neovascularization. We investigated whether transplantation of mouse adipose tissue-derived stem cells (mADSC) into the intraperitoneal space has a rescue effect on NaIO3 -induced retinal degeneration in mice. In this study, mADSC transplantation recovered visual function and preserved the retinal outer layer structure compared to the control group without any integration of mADSC into the retina. Moreover, endogenous ciliary neurotrophic factor (CNTF) was elevated in the retinas of mADSC-treated mice. We found that lipopolysaccharide (LPS) or LPS-stimulated monocyte supernatant induced the secretion of granulocyte colony stimulating factor (GCSF), CD54, CXCL10, interleukin-6 (IL-6), and CCL5 from the mADSC by cytokine array. Network inference was conducted to investigate signaling networks related to CNTF regulation. Based on bioinformatics data, the expression of IL-6 was related to the expression of CNTF. Additionally, intravitreal injection of IL-6 in rats produced up-regulation of endogenous CNTF in the retina. mADSC had a rescue effect on retinal degeneration through the up-regulation of endogenous CNTF by IL-6. Thus, transplantation of mADSC could be a potential treatment option for retinal degeneration.
Collapse
Affiliation(s)
- Jeong Hoon Heo
- Department of Molecular Biology and Immunology, College of Medicine, Kosin University, Pusan, Korea
- Institute for Medicine, College of Medicine, Kosin University, Pusan, Korea
| | - Jung Ae Yoon
- Department of Dental Hygiene, Dong Ju College, Pusan, Korea
| | - Eun Kyung Ahn
- Department of Biological Science, College of Natural Science, Dong-A University, Pusan, Korea
| | - Hyun Kim
- Department of Anatomy, College of Medicine, Kosin University, Pusan, Korea
| | - Sang Hwa Urm
- Department of Preventive Medicine, Inje University College of Medicine, Pusan, Korea
| | - Chul Oh Oak
- Department of Internal Medicine, College of Medicine, Kosin University, Pusan, Korea
| | - Byeng Chul Yu
- Department of Preventive Medicine, College of Medicine, Kosin University, Pusan, Korea
| | - Sang Joon Lee
- Institute for Medicine, College of Medicine, Kosin University, Pusan, Korea
- Department of Ophthalmology, College of Medicine, Kosin University, Pusan, Korea
| |
Collapse
|
20
|
Hurst J, Vitkute M, Hofmann K, Müller S, Löscher M, Bartz-Schmidt KU, Spitzer MS, Schnichels S, Januschowski K. Comparison of Different Cell Culture Media in the Model of the Isolated and Superfused Bovine Retina: Investigating the Limits of More Physiological Perfusion Solutions. Curr Eye Res 2017; 43:232-243. [PMID: 29111831 DOI: 10.1080/02713683.2017.1387668] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
PURPOSE The isolated superfused retina is a standardized tool in ophthalmological research. However, stable electroretinogram (ERG) responses can only be obtained for around eight hours; therefore, limiting its use. The aim of this study was to evaluate the short-term potential of different cell culture media and to promote long-term testing based on the results obtained. MATERIALS AND METHODS For the experimental procedure bovine retinae were prepared and perfused with the standard Sickel solution and an ERG was performed. After recording stable a- or b-waves, different media (Dulbecco's Modified Eagle's Medium (DMEM), MACS, and Neurobasal) were superfused for 45 minutes. ERG recovery was monitored overall for 75 minutes. Analysis of the mRNA expression of Thy-1, GFAP, Bax/Bcl-2-ratio, Rhodopsin, and Opsin via qRT-PCR was performed directly after ERG recording on the same retina. RESULTS None of the tested media had a negative effect on a-wave amplitudes, although b-wave amplitudes decreased (DMEM) or increased (MACS and Neurobasal) compared to the standard solution (Sickel) after 45 minutes of exposure. However, after 75 minutes of wash-out, no difference to the standard solution alone could be observed. Exposure to different media either had no effect or decreased the Opsin and Rhodopsin mRNA levels. Thy-1 expression was strongly diminished in DMEM and MACS (by 2-3-fold), whereas incubation in Neurobasal medium led to a slight increase compared to incubation with the standard solution. Furthermore, the Bax/Bcl-2 ratio indicated an anti-apoptotic effect (Bax/Bcl-2 = 0.16; p < 0.05) for Neurobasal. CONCLUSION Neurobasal medium displayed the best electrophysiological properties in the short-term and may be applicable for stable long-term escalation testing.
Collapse
Affiliation(s)
- José Hurst
- a Centre for Ophthalmology, University Eye Hospital Tübingen , Tübingen , Germany
| | - Milda Vitkute
- a Centre for Ophthalmology, University Eye Hospital Tübingen , Tübingen , Germany
| | - Kathleen Hofmann
- a Centre for Ophthalmology, University Eye Hospital Tübingen , Tübingen , Germany
| | - Sebastian Müller
- a Centre for Ophthalmology, University Eye Hospital Tübingen , Tübingen , Germany
| | - Marina Löscher
- a Centre for Ophthalmology, University Eye Hospital Tübingen , Tübingen , Germany
| | | | - Martin S Spitzer
- a Centre for Ophthalmology, University Eye Hospital Tübingen , Tübingen , Germany.,b Department of Ophthalmology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sven Schnichels
- a Centre for Ophthalmology, University Eye Hospital Tübingen , Tübingen , Germany
| | - Kai Januschowski
- a Centre for Ophthalmology, University Eye Hospital Tübingen , Tübingen , Germany.,c Eye Hospital , Sulzbach , Saar , Germany
| |
Collapse
|
21
|
Ghasemi M, Alizadeh E, Saei Arezoumand K, Fallahi Motlagh B, Zarghami N. Ciliary neurotrophic factor (CNTF) delivery to retina: an overview of current research advancements. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:1694-1707. [PMID: 29065723 DOI: 10.1080/21691401.2017.1391820] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The intraocular administration of the ciliary neurotrophic factor (CNTF) has been found to attenuate the photoreceptor degeneration and preserve retinal functions in the animal research models of the inherited or induced retinal disease. Studies with the aim of CNTF transfer to the posterior segment inside the eye have been directed to determine the best method for its administration. An ideal delivery method would overcome the eye drug elimination mechanisms or barriers and provide the sustained release of the CNTF into retina in the safest fashion with the minimum harm to the quality of life. This review focuses on the present state of CNTF delivery to retina, also provides an overview of available technologies and their challenges.
Collapse
Affiliation(s)
- Maryam Ghasemi
- a The Umbilical Cord Stem Cell Research Center (UCSRC) , Tabriz University of Medical Sciences , Tabriz , Iran.,b Department of Medical Biotechnology, Faculty of Advanced Medical Sciences , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Effat Alizadeh
- a The Umbilical Cord Stem Cell Research Center (UCSRC) , Tabriz University of Medical Sciences , Tabriz , Iran.,b Department of Medical Biotechnology, Faculty of Advanced Medical Sciences , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Khatereh Saei Arezoumand
- b Department of Medical Biotechnology, Faculty of Advanced Medical Sciences , Tabriz University of Medical Sciences , Tabriz , Iran
| | | | - Nosratollah Zarghami
- a The Umbilical Cord Stem Cell Research Center (UCSRC) , Tabriz University of Medical Sciences , Tabriz , Iran.,b Department of Medical Biotechnology, Faculty of Advanced Medical Sciences , Tabriz University of Medical Sciences , Tabriz , Iran.,d Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine , Tabriz University of Medical Sciences , Tabriz , Iran
| |
Collapse
|
22
|
Affiliation(s)
- Gustavo D. Aguirre
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| |
Collapse
|
23
|
Protective effect of clusterin on rod photoreceptor in rat model of retinitis pigmentosa. PLoS One 2017; 12:e0182389. [PMID: 28767729 PMCID: PMC5540409 DOI: 10.1371/journal.pone.0182389] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 07/17/2017] [Indexed: 01/09/2023] Open
Abstract
Retinitis Pigmentosa (RP) begins with the death of rod photoreceptors and is slowly followed by a gradual loss of cones and a rearrangement of the remaining retinal neurons. Clusterin is a chaperone protein that protects cells and is involved in various pathophysiological stresses, including retinal degeneration. Using a well-established transgenic rat model of RP (rhodopsin S334ter), we investigated the effects of clusterin on rod photoreceptor survival. To investigate the role of clusterin in S334ter-line3 retinas, Voronoi analysis and immunohistochemistry were used to evaluate the geometry of rod distribution. Additionally, immunoblot analysis, Bax activation, STAT3 and Akt phosphorylation were used to evaluate the pathway involved in rod cell protection. In this study, clusterin (10μg/ml) intravitreal treatment produced robust preservation of rod photoreceptors in S334ter-line3 retina. The mean number of rods in 1mm2 was significantly greater in clusterin injected RP retinas (postnatal (P) 30, P45, P60, & P75) than in age-matched saline injected RP retinas (P<0.01). Clusterin activated Akt, STAT3 and significantly reduced Bax activity; in addition to inducing phosphorylated STAT3 in Müller cells, which suggests it may indirectly acts on photoreceptors. Thus, clusterin treatment may interferes with mechanisms leading to rod death by suppressing cell death through activation of Akt and STAT3, followed by Bax suppression. Novel insights into the pathway of how clusterin promotes the rod cell survival suggest this treatment may be a potential therapeutic strategy to slow progression of vision loss in human RP.
Collapse
|
24
|
Zhou WD, Wang LL, Zhou LB, Bin W, Bao TP, Zhang Y, Shu J, Yang WX, Hui LL, Jin R, Zhuang LL, Zhou GP. All-trans retinoic acid upregulates the expression of ciliary neurotrophic factor in retinal pigment epithelial cells. Cell Biochem Funct 2017; 35:202-208. [PMID: 28589680 DOI: 10.1002/cbf.3264] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 03/07/2017] [Accepted: 03/13/2017] [Indexed: 11/10/2022]
Abstract
Retinopathy of prematurity, a leading cause of visual impairment in low birth-weight infants, remains a crucial therapeutic challenge. Ciliary neurotrophic factor (CNTF) is a promyelinating trophic factor that promotes rod and cone photoreceptor survival and cone outer segment regeneration in the degenerating retina. Ciliary neurotrophic factor expression is regulated by many factors such as all-trans retinoic acid (ATRA). In this study, we found that ATRA increased CNTF expression in mouse retinal pigment epithelial (RPE) cells in a dose- and time-dependent manner, and PKA signaling pathway is necessary for ATRA-induced CNTF upregulation. Furthermore, we showed that ATRA promoted CNTF expression through CREB binding to its promoter region. In addition, CNTF levels were decreased in serum of retinopathy of prematurity children and in retinal tissue of oxygen-induced retinopathy mice. In mouse RPE cells cultured with high oxygen, CNTF expression and secretion were decreased, but could be recovered after treatment with ATRA. In conclusion, our data suggest that ATRA administration upregulates CNTF expression in RPE cells.
Collapse
Affiliation(s)
- Wen-Di Zhou
- Department of Pediatrics, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China.,Department of Pediatrics, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Lu-Lu Wang
- Department of Pediatrics, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Lan-Bo Zhou
- 2013 Clinical Class 7, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, P.R., China
| | - Wei Bin
- Department of Pediatrics, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Tian-Ping Bao
- Department of Pediatrics, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Yi Zhang
- Department of Ophthalmology, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jin Shu
- Department of Pediatrics, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Wei-Xia Yang
- Department of Pediatrics, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Liang-Liang Hui
- Department of Pediatrics, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Rui Jin
- Department of Pediatrics, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Li-Li Zhuang
- Department of Pediatrics, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Guo-Ping Zhou
- Department of Pediatrics, The First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
25
|
Significant changes in endogenous retinal gene expression assessed 1 year after a single intraocular injection of AAV-CNTF or AAV-BDNF. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 3:16078. [PMID: 27933306 PMCID: PMC5142514 DOI: 10.1038/mtm.2016.78] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Revised: 09/27/2016] [Accepted: 10/12/2016] [Indexed: 12/17/2022]
Abstract
Use of viral vectors to deliver therapeutic genes to the central nervous system holds promise for the treatment of neurodegenerative diseases and neurotrauma. Adeno-associated viral (AAV) vectors encoding brain-derived neurotrophic factor (BDNF) or ciliary derived neurotrophic factor (CNTF) promote the viability and regeneration of injured adult rat retinal ganglion cells. However, these growth-inducing transgenes are driven by a constitutively active promoter, thus we examined whether long-term AAV-mediated secretion of BDNF or CNTF affected endogenous retinal gene expression. One year after the intravitreal injection of AAV-green fluorescent protein (GFP), bi-cistronic AAV-BDNF-GFP or AAV-CNTF-GFP, mRNA was extracted and analyzed using custom 96 well polymerase chain reaction arrays. Of 93 test genes, 56% showed significantly altered expression in AAV-BDNF-GFP and/or AAV-CNTF-GFP retinas compared with AAV-GFP controls. Of these genes, 73% showed differential expression in AAV-BDNF versus AAV-CNTF injected eyes. To focus on retinal ganglion cell changes, quantitative polymerase chain reaction was undertaken on mRNA (16 genes) obtained from fixed retinal sections in which the ganglion cell layer was enriched. The sign and extent of fold changes in ganglion cell layer gene expression differed markedly from whole retinal samples. Sustained and global alteration in endogenous mRNA expression after gene therapy should be factored into any interpretation of experimental/clinical outcomes, particularly when introducing factors into the central nervous system that require secretion to evoke functionality.
Collapse
|
26
|
Abstract
Over the last few years, huge progress has been made with regard to the understanding of molecular mechanisms underlying the pathogenesis of neurodegenerative diseases of the eye. Such knowledge has led to the development of gene therapy approaches to treat these devastating disorders. Challenges regarding the efficacy and efficiency of therapeutic gene delivery have driven the development of novel therapeutic approaches, which continue to evolve the field of ocular gene therapy. In this review article, we will discuss the evolution of preclinical and clinical strategies that have improved gene therapy in the eye, showing that treatment of vision loss has a bright future.
Collapse
Affiliation(s)
- Lolita Petit
- 1 Department of Ophthalmology and Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Hemant Khanna
- 1 Department of Ophthalmology and Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Department of Neurobiology, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Claudio Punzo
- 1 Department of Ophthalmology and Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Department of Neurobiology, University of Massachusetts Medical School , Worcester, Massachusetts
| |
Collapse
|
27
|
Marangoni D, Vijayasarathy C, Bush RA, Wei LL, Wen R, Sieving PA. Intravitreal Ciliary Neurotrophic Factor Transiently Improves Cone-Mediated Function in a CNGB3-/- Mouse Model of Achromatopsia. Invest Ophthalmol Vis Sci 2016; 56:6810-22. [PMID: 26567794 DOI: 10.1167/iovs.15-16866] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Ciliary neurotrophic factor (CNTF) was recently shown to augment cone function in CNGB3 mutant achromat dogs. However, testing CNTF-releasing implant in human CNGB3 achromats failed to show benefit. We evaluated the effects of CNTF protein on the retinal function in an additional achromatopsia model, the CNGB3-/- mouse. METHODS Fifty-nine CNGB3-/- mice (postnatal day [PD] ± SD = 30 ± 7) received a unilateral intravitreal injection of 1 or 2 μg CNTF protein, and 15 wild-type (WT) mice (PD = 34 ± 3) received 1 μg CNTF. Retinal function was evaluated by flash ERG and photopic flicker ERG (fERG) at 7 and 14 days after treatment. RESULTS Seven days post CNTF, the photopic b-wave Vmax was significantly increased in CNGB3-/- mice (P < 0.01), whereas it was reduced in WT mice (P < 0.05). Ciliary neurotrophic factor significantly increased the amplitude of photopic fERG and the photopic oscillatory potentials (OPs) in CNGB3-/- mice. Ciliary neurotrophic factor did not alter the scotopic a-wave in either CNGB3-/- or WT mice, but it increased the scotopic b-wave k (P < 0.01) in CNGB3-/- mice, indicating diminished scotopic sensitivity, and reduced the scotopic b-wave Vmax in WT mice (P < 0.05). No difference was found in ERG parameters between 1 or 2 μg CNTF. Fourteen days after CNTF injection the ERG changes in CNGB3-/- mice were lost. CONCLUSIONS Intravitreal bolus CNTF protein caused a small and transient improvement of cone-mediated function in CNGB3-/- mice, whereas it reduced rod-mediated function. The increase in photopic OPs and the lack of changes in scotopic a-wave suggest a CNTF effect on the inner retina.
Collapse
Affiliation(s)
- Dario Marangoni
- National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, United States 3Department of Biotechnology and Applied Clinical Science, University of L'Aquila, L'Aquila, Italy
| | - Camasamudram Vijayasarathy
- National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, United States
| | - Ronald A Bush
- National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, United States
| | - Lisa L Wei
- National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Rong Wen
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami, Miller School of Medicine, Miami, Florida, United States
| | - Paul A Sieving
- National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, United States 2National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
28
|
Song D, Wilson B, Zhao L, Bhuyan R, Bandyopadhyay M, Lyubarsky A, Yu C, Li Y, Kanu L, Miwa T, Song WC, Finnemann SC, Rohrer B, Dunaief JL. Retinal Pre-Conditioning by CD59a Knockout Protects against Light-Induced Photoreceptor Degeneration. PLoS One 2016; 11:e0166348. [PMID: 27893831 PMCID: PMC5125596 DOI: 10.1371/journal.pone.0166348] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 10/27/2016] [Indexed: 11/25/2022] Open
Abstract
Complement dysregulation plays a key role in the pathogenesis of age-related macular degeneration (AMD), but the specific mechanisms are incompletely understood. Complement also potentiates retinal degeneration in the murine light damage model. To test the retinal function of CD59a, a complement inhibitor, CD59a knockout (KO) mice were used for light damage (LD) experiments. Retinal degeneration and function were compared in WT versus KO mice following light damage. Gene expression changes, endoplasmic reticulum (ER) stress, and glial cell activation were also compared. At baseline, the ERG responses and rhodopsin levels were lower in CD59aKO compared to wild-type (WT) mice. Following LD, the ERG responses were better preserved in CD59aKO compared to WT mice. Correspondingly, the number of photoreceptors was higher in CD59aKO retinas than WT controls after LD. Under normal light conditions, CD59aKO mice had higher levels than WT for GFAP immunostaining in Müller cells, mRNA and protein levels of two ER-stress markers, and neurotrophic factors. The reduction in photon capture, together with the neurotrophic factor upregulation, may explain the structural and functional protection against LD in the CD59aKO.
Collapse
Affiliation(s)
- Delu Song
- The F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA
| | - Brooks Wilson
- Department of Ophthalmology, Medical University of South Carolina, Charleston, SC
| | - Liangliang Zhao
- The F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA
- Department of Ophthalmology, The Second Hospital of Jilin University, Jilin, China
| | - Rupak Bhuyan
- The F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA
| | | | - Arkady Lyubarsky
- The F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA
| | - Chen Yu
- Center for Cancer, Genetic Diseases, and Gene Regulation, Department of Biological Sciences, Fordham University, Bronx, NY
| | - Yafeng Li
- The F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA
| | - Levi Kanu
- The F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA
| | - Takashi Miwa
- Department of Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Wen-Chao Song
- Department of Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Silvia C. Finnemann
- Center for Cancer, Genetic Diseases, and Gene Regulation, Department of Biological Sciences, Fordham University, Bronx, NY
| | - Bärbel Rohrer
- Department of Ophthalmology, Medical University of South Carolina, Charleston, SC
- Research Service, Ralph H. Johnson VA Medical Center, Charleston, SC
- * E-mail: (JLD); (BR)
| | - Joshua L. Dunaief
- The F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA
- * E-mail: (JLD); (BR)
| |
Collapse
|
29
|
Guadagni V, Novelli E, Piano I, Gargini C, Strettoi E. Pharmacological approaches to retinitis pigmentosa: A laboratory perspective. Prog Retin Eye Res 2015; 48:62-81. [PMID: 26113212 DOI: 10.1016/j.preteyeres.2015.06.005] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 06/05/2015] [Accepted: 06/11/2015] [Indexed: 01/08/2023]
Abstract
Retinal photoreceptors are highly specialized and performing neurons. Their cellular architecture is exquisitely designed to host a high concentration of molecules involved in light capture, phototransduction, electric and chemical signaling, membrane and molecular turnover, light and dark adaption, network activities etc. Such high efficiency and molecular complexity require a great metabolic demand, altogether conferring to photoreceptors particular susceptibility to external and internal insults, whose occurrence usually precipitate into degeneration of these cells and blindness. In Retinitis Pigmentosa, an impressive number of mutations in genes expressed in the retina and coding for a large varieties of proteins leads to the progressive death of photoreceptors and blindness. Recent advances in molecular tools have greatly facilitated the identification of the underlying genetics and molecular bases of RP leading to the successful implementation of gene therapy for some types of mutations, with visual restoration in human patients. Yet, genetic heterogeneity of RP makes mutation-independent approaches highly desirable, although many obstacles pave the way to general strategies for treating this complex disease, which remains orphan. The review will focus on treatments for RP based on pharmacological tools, choosing, among the many ongoing studies, approaches which rely on strong experimental evidence or rationale. For perspective treatments, new concepts are foreseen to emerge from basic studies elucidating the pathways connecting the primary mutations to photoreceptor death, possibly revealing common molecular targets for drug intervention.
Collapse
Affiliation(s)
- Viviana Guadagni
- Neuroscience Institute, Italian National Research Council (CNR), Area della Ricerca, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy
| | - Elena Novelli
- Neuroscience Institute, Italian National Research Council (CNR), Area della Ricerca, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy
| | - Ilaria Piano
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | | | - Enrica Strettoi
- Neuroscience Institute, Italian National Research Council (CNR), Area della Ricerca, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy.
| |
Collapse
|
30
|
Jankowiak W, Kruszewski K, Flachsbarth K, Skevas C, Richard G, Rüther K, Braulke T, Bartsch U. Sustained Neural Stem Cell-Based Intraocular Delivery of CNTF Attenuates Photoreceptor Loss in the nclf Mouse Model of Neuronal Ceroid Lipofuscinosis. PLoS One 2015; 10:e0127204. [PMID: 25992714 PMCID: PMC4439090 DOI: 10.1371/journal.pone.0127204] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 04/13/2015] [Indexed: 01/10/2023] Open
Abstract
A sustained intraocular administration of neurotrophic factors is among the strategies aimed at establishing treatments for currently untreatable degenerative retinal disorders. In the present study we have analyzed the neuroprotective effects of a continuous neural stem (NS) cell-based intraocular delivery of ciliary neurotrophic factor (CNTF) on photoreceptor cells in the nclf mouse, an animal model of the neurodegenerative lysosomal storage disorder variant late infantile neuronal ceroid lipofuscinosis (vLINCL). To this aim, we genetically modified adherently cultivated NS cells with a polycistronic lentiviral vector encoding a secretable variant of CNTF together with a Venus reporter gene (CNTF-NS cells). NS cells for control experiments (control-NS cells) were modified with a vector encoding the reporter gene tdTomato. Clonal CNTF-NS and control-NS cell lines were established using fluorescent activated cell sorting and intravitreally grafted into 14 days old nclf mice at the onset of retinal degeneration. The grafted cells preferentially differentiated into astrocytes that were attached to the posterior side of the lenses and the vitreal side of the retinas and stably expressed the transgenes for at least six weeks, the latest post-transplantation time point analyzed. Integration of donor cells into host retinas, ongoing proliferation of grafted cells or adverse effects of the donor cells on the morphology of the host eyes were not observed. Quantitative analyses of host retinas two, four and six weeks after cell transplantation revealed the presence of significantly more photoreceptor cells in eyes with grafted CNTF-NS cells than in eyes with grafted control-NS cells. This is the first demonstration that a continuous intraocular administration of a neurotrophic factor attenuates retinal degeneration in an animal model of neuronal ceroid lipofuscinosis.
Collapse
Affiliation(s)
- Wanda Jankowiak
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katharina Kruszewski
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kai Flachsbarth
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christos Skevas
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gisbert Richard
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Klaus Rüther
- Department of Ophthalmology, Sankt Gertrauden-Krankenhaus, Berlin, Germany
| | - Thomas Braulke
- Department of Biochemistry, Children’s Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Udo Bartsch
- Department of Ophthalmology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- * E-mail:
| |
Collapse
|
31
|
Lipinski DM, Barnard AR, Singh MS, Martin C, Lee EJ, Davies WIL, MacLaren RE. CNTF Gene Therapy Confers Lifelong Neuroprotection in a Mouse Model of Human Retinitis Pigmentosa. Mol Ther 2015; 23:1308-1319. [PMID: 25896245 DOI: 10.1038/mt.2015.68] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 02/28/2015] [Indexed: 12/12/2022] Open
Abstract
The long-term outcome of neuroprotection as a therapeutic strategy for preventing cell death in neurodegenerative disorders remains unknown, primarily due to slow disease progression and the inherent difficulty of assessing neuronal survival in vivo. Employing a murine model of retinal disease, we demonstrate that ciliary neurotrophic factor (CNTF) confers life-long protection against photoreceptor degeneration. Repetitive retinal imaging allowed the survival of intrinsically fluorescent cone photoreceptors to be quantified in vivo. Imaging of the visual cortex and assessment of visually-evoked behavioral responses demonstrated that surviving cones retain function and signal correctly to the brain. The mechanisms underlying CNTF-mediated neuroprotection were explored through transcriptome analysis, revealing widespread upregulation of proteolysis inhibitors, which may prevent cellular/extracellular matrix degradation and complement activation in neurodegenerative diseases. These findings provide insights into potential novel therapeutic avenues for diseases such as retinitis pigmentosa and amyotrophic lateral sclerosis, for which CNTF has been evaluated unsuccessfully in clinical trials.
Collapse
Affiliation(s)
- Daniel M Lipinski
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; Oxford Biomedical Research Centre, University of Oxford, Oxford, UK; Department of Ophthalmology, University of Florida, Gainesville, Florida, USA
| | - Alun R Barnard
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Mandeep S Singh
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Chris Martin
- Department of Psychology, The University of Sheffield, Sheffield, UK
| | - Edward J Lee
- Moorfields Eye Hospital & UCL NIHR Biomedical Research Centre for Ophthalmology, London, UK
| | - Wayne I L Davies
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; School of Animal Biology and University of Western Australia Oceans Institute, University of Western Australia, Perth, Australia
| | - Robert E MacLaren
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; Oxford Biomedical Research Centre, University of Oxford, Oxford, UK; Moorfields Eye Hospital & UCL NIHR Biomedical Research Centre for Ophthalmology, London, UK.
| |
Collapse
|
32
|
Chew EY, Clemons TE, Peto T, Sallo FB, Ingerman A, Tao W, Singerman L, Schwartz SD, Peachey NS, Bird AC. Ciliary neurotrophic factor for macular telangiectasia type 2: results from a phase 1 safety trial. Am J Ophthalmol 2015; 159:659-666.e1. [PMID: 25528956 DOI: 10.1016/j.ajo.2014.12.013] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Revised: 12/10/2014] [Accepted: 12/11/2014] [Indexed: 11/18/2022]
Abstract
PURPOSE To evaluate the safety and tolerability of intraocular delivery of ciliary neurotrophic factor (CNTF) using an encapsulated cell implant for the treatment of macular telangiectasia type 2. DESIGN An open-label safety trial conducted in 2 centers enrolling 7 participants with macular telangiectasia type 2. METHODS The participant's more severely affected eye (worse baseline visual acuity) received the high-dose implant of CNTF. Patients were followed for a period of 36 months. The primary safety outcome was a change in the parameters of the electroretinogram (ERG). Secondary efficacy outcomes were changes in visual acuity, en face measurements of the optical coherence tomography of the disruption in the ellipsoid zone, and microperimetry when compared with baseline. RESULTS The ERG findings demonstrated a reduction in the amplitude of the scotopic b-wave in 4 participants 3 months after implantation (month 3). All parameters returned to baseline values by month 12 and remained so at month 36 with no clinical impact on dark adaptation. There was no change in visual acuity compared with baseline. The area of the defect as measured functionally by microperimetry and structurally by the en face OCT imaging of the ellipsoid zone loss appeared unchanged from baseline. CONCLUSIONS The intraocular delivery of CNTF in the encapsulated cell implant appeared to be safe and well tolerated in eyes with macular telangiectasia type 2. Further evaluation in a randomized controlled clinical trial is warranted to test for efficacy.
Collapse
Affiliation(s)
- Emily Y Chew
- National Eye Institute, National Institutes of Health, Bethesda, Maryland.
| | | | - Tunde Peto
- National Institute of Health Research Biomedical Research Centre at Moorfields Eye Hospital National Health Service Foundation Trust and UCL Institute of Ophthalmology, London, United Kingdom
| | - Ferenc B Sallo
- Department of Research and Development, Moorfields Eye Hospital, London, United Kingdom; University College London, Institute of Ophthalmology, London, United Kingdom
| | | | - Weng Tao
- Neurotech Pharmaceuticals, Cumberland, Rhode Island
| | | | | | - Neal S Peachey
- Cleveland Clinic, Department of Ophthalmology, Cleveland Ohio; Louis Stokes Cleveland VA Medical Center, Cleveland, Ohio
| | - Alan C Bird
- Department of Inherited Eye Disease, Moorfields Eye Hospital, London, United Kingdom
| |
Collapse
|
33
|
Petersen-Jones SM, Komáromy AM. Dog models for blinding inherited retinal dystrophies. HUM GENE THER CL DEV 2015; 26:15-26. [PMID: 25671556 DOI: 10.1089/humc.2014.155] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Spontaneous canine models exist for several inherited retinal dystrophies. This review will summarize the models and indicate where they have been used in translational gene therapy trials. The RPE65 gene therapy trials to treat childhood blindness are a good example of how studies in dogs have contributed to therapy development. Outcomes in human clinical trials are compared and contrasted with the result of the preclinical dog trials.
Collapse
Affiliation(s)
- Simon M Petersen-Jones
- 1 Department of Small Animal Clinical Sciences, Michigan State University , East Lansing, MI 48824
| | | |
Collapse
|
34
|
Petersen-Jones S, Komaromy AM. Dog Models for Blinding Inherited Retinal Degenerations. HUM GENE THER CL DEV 2014. [DOI: 10.1089/hum.2014.155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
35
|
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
|
36
|
Xia X, Wen R, Chou TH, Li Y, Wang Z, Porciatti V. Protection of pattern electroretinogram and retinal ganglion cells by oncostatin M after optic nerve injury. PLoS One 2014; 9:e108524. [PMID: 25243471 PMCID: PMC4171539 DOI: 10.1371/journal.pone.0108524] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 09/01/2014] [Indexed: 12/25/2022] Open
Abstract
Injury to retinal ganglion cell (RGC) axons leads to selective loss of RGCs and vision. Previous studies have shown that exogenous neurotrophic factors promote RGC survival. We investigated the neuroprotective effects of oncostatin M (OSM), a member of the IL-6 family of cytokines, on pattern electroretinogram (PERG) and RGC survival after optic nerve crush (ON-crush) in the mouse. BALB/C mice received ON-crush in the left eyes for either 4-second or 1-second duration (4-s or 1-s). Fluoro-gold retrograde labeling was used to identify RGCs. RGC function was assessed by PERG measurement. OSM or CNTF protein was injected intravitreally immediately after ON-crush. OSM responsive cells were identified by localization of increased STAT3 phosphorylation. Significant higher RGC survival (46% of untreated control) was seen in OSM-treated eyes when assessed 2 weeks after 4-s ON-crush as compared to that (14% of untreated control) of the PBS-treated eyes (P<0.001). In addition, PERG amplitude was significantly higher in eyes treated with OSM or CNTF 1 week after 1-s ON-crush (36% of baseline) as compared with the amplitude of PBS-treated eyes (19% of the baseline, P = 0.003). An increase in STAT3 phosphorylation was localized in Müller layer after OSM treatment, suggesting that Müller cells mediate the effect of OSM. Our results demonstrate that one single injection of either OSM or CNTF after ON-crush improves RGC survival together with their electrophysiological activity. These data provide proof-of-concept for using neurotrophic factors OSM and CNTF for RGC degenerative diseases, including glaucoma and acute optic nerve trauma.
Collapse
Affiliation(s)
- Xin Xia
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
- Department of Ophthalmology, Shanghai First People’s Hospital, Jiaotong University, Shanghai, China
- Shanghai Key Laboratory for Ocular Fundus Diseases, Shanghai, China
| | - Rong Wen
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
| | - Tsung-Han Chou
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
| | - Yiwen Li
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
| | - Zhengying Wang
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
| | - Vittorio Porciatti
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
- * E-mail:
| |
Collapse
|
37
|
Zein WM, Jeffrey BG, Wiley HE, Turriff AE, Tumminia SJ, Tao W, Bush RA, Marangoni D, Wen R, Wei LL, Sieving PA. CNGB3-achromatopsia clinical trial with CNTF: diminished rod pathway responses with no evidence of improvement in cone function. Invest Ophthalmol Vis Sci 2014; 55:6301-8. [PMID: 25205868 DOI: 10.1167/iovs.14-14860] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Ciliary neurotrophic factor (CNTF) protects rod photoreceptors from retinal degenerative disease in multiple nonhuman models. Thus far, CNTF has failed to demonstrate rod protection in trials for human retinitis pigmentosa. Recently, CNTF was found to improve cone photoreceptor function in a canine CNGB3 achromatopsia model. This study explores whether this finding translates to humans with CNGB3 achromatopsia. METHODS A five-subject, open-label Phase I/II study was initiated by implanting intraocular microcapsules releasing CNTF (nominally 20 ng/d) into one eye each of CNGB3 achromat participants. Fellow eyes served as untreated controls. Subjects were followed for 1 year. RESULTS Pupil constriction in treated eyes gave evidence of intraocular CNTF release. Additionally, scotopic ERG responses were reduced, and dark-adapted psychophysical absolute thresholds were increased, attributable to diminished rod or rod pathway activity. Optical coherence tomography revealed that the cone-rich fovea underwent structural changes as the foveal hyporeflective zone (HRZ) became diminished in CNTF-treated eyes. No objectively measurable enhancement of cone function was found by assessments of visual acuity, mesopic increment sensitivity threshold, or the photopic ERG. Careful measurements of color hue discrimination showed no change. Nonetheless, subjects reported beneficial changes of visual function in the treated eyes, including reduced light sensitivity and aversion to bright light, which may trace to decreased effective ambient light from the pupillary constriction; further they noted slowed adaptation to darkness, consistent with CNTF action on rod photoreceptors. CONCLUSIONS Ciliary neurotrophic factor did not measurably enhance cone function, which reveals a species difference between human and canine CNGB3 cones in response to CNTF. (ClinicalTrials.gov number, NCT01648452.).
Collapse
Affiliation(s)
- Wadih M Zein
- National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Brett G Jeffrey
- National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Henry E Wiley
- National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Amy E Turriff
- National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Santa J Tumminia
- National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Weng Tao
- Neurotech Pharmaceuticals, Inc., Cumberland, Rhode Island, United States
| | - Ronald A Bush
- National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, United States
| | - Dario Marangoni
- National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, United States Department of Biotechnology and Applied Clinical Science, University of L'Aquila, L'Aquila, Italy
| | - Rong Wen
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami, Miller School of Medicine, Miami, Florida, United States
| | - Lisa L Wei
- National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Paul A Sieving
- National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
38
|
Kucharska J, Del Río P, Arango-Gonzalez B, Gorza M, Feuchtinger A, Hauck SM, Ueffing M. Cyr61 activates retinal cells and prolongs photoreceptor survival in rd1 mouse model of retinitis pigmentosa. J Neurochem 2014; 130:227-40. [PMID: 24593181 DOI: 10.1111/jnc.12704] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 02/27/2014] [Accepted: 02/27/2014] [Indexed: 01/08/2023]
Abstract
Subretinal injections with glial cell line-derived neurotrophic factor (GDNF) rescue morphology as well as function of rod cells in mouse and rat animal models of retinitis pigmentosa. At the same time, it is postulated that this effect is indirect, mediated by activation of retinal Müller glial (RMG) cells. Here, we show that Cyr61/CCN1, one of the secreted proteins up-regulated in primary RMG after glial cell line-derived neurotrophic factor stimulation, provides neuroprotective and pro-survival capacities: Recombinant Cyr61 significantly reduced photoreceptor (PR) cells death in organotypic cultures of Pde6b(rd1) retinas. To identify stimulated pathways in the retina, we treated Pde6b(rd1) retinal explants with Cyr61 and observed an overall increase in activated Erk1/2 and Stat3 signalling molecules characterized by activation-site-specific phosphorylation. To identify Cyr61 retinal target cells, we isolated primary porcine PR, RMG and retinal pigment epithelium (RPE) cells and exposed them separately to Cyr61. Here, RMG as well as RPE cells responded with induced phosphorylation of Erk1/2, Stat3 and Akt. In PR, no increase in phosphorylation in any of the studied proteins was detected, suggesting an indirect neuroprotective effect of Cyr61. Cyr61 may thus act as an endogenous pro-survival factor for PR, contributing to the complex repertoire of neuroprotective activities generated by RMG and RPE cells. We propose the following model of Cyr61 neuroprotection within the retina: Cyr61 stimulates retinal Müller glial (RMG) and retinal pigment epithelium (RPE) cells and activates PI3K/Akt, mitogen-activated protein kinase(MAPK)/Erk and Janus kinase(JAK)/Stat-signalling pathways in these cells. Phosphorylated Stat3 and Erk1/2 presumably translocate to the nucleus, induce transcriptional changes, which increase secretion of neuroprotective agents that protect photoreceptors (PR) from mutation-induced death.
Collapse
Affiliation(s)
- Joanna Kucharska
- Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany; Research Unit Protein Science, Helmholtz Zentrum München, Neuherberg, Germany
| | | | | | | | | | | | | |
Collapse
|
39
|
CNTF-mediated protection of photoreceptors requires initial activation of the cytokine receptor gp130 in Müller glial cells. Proc Natl Acad Sci U S A 2013; 110:E4520-9. [PMID: 24191003 DOI: 10.1073/pnas.1303604110] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Ciliary neurotrophic factor (CNTF) acts as a potent neuroprotective agent in multiple retinal degeneration animal models. Recently, CNTF has been evaluated in clinical trials for the inherited degenerative disease retinitis pigmentosa (RP) and for dry age-related macular degeneration (AMD). Despite its potential as a broad-spectrum therapeutic treatment for blinding diseases, the target cells of exogenous CNTF and its mechanism of action remain poorly understood. We have shown previously that constitutive expression of CNTF prevents photoreceptor death but alters the retinal transcriptome and suppresses visual function. Here, we use a lentivirus to deliver the same secreted human CNTF used in clinical trials to a mouse model of RP. We found that low levels of CNTF halt photoreceptor death, improve photoreceptor morphology, and correct opsin mislocalization. However, we did not detect corresponding improvement of retinal function as measured by the electroretinogram. Disruption of the cytokine receptor gp130 gene in Müller glia reduces CNTF-dependent photoreceptor survival and prevents phosphorylation of STAT3 and ERK in Müller glia and the rest of the retina. Targeted deletion of gp130 in rods also demolishes neuroprotection by CNTF and prevents further activation of Müller glia. Moreover, CNTF elevates the expression of LIF and endothelin 2, thus positively promoting Müller and photoreceptor interactions. We propose that exogenous CNTF initially targets Müller glia, and subsequently induces cytokines acting through gp130 in photoreceptors to promote neuronal survival. These results elucidate a cellular mechanism for exogenous CNTF-triggered neuroprotection and provide insight into the complex cellular responses induced by CNTF in diseased retinas.
Collapse
|
40
|
Jung G, Sun J, Petrowitz B, Riecken K, Kruszewski K, Jankowiak W, Kunst F, Skevas C, Richard G, Fehse B, Bartsch U. Genetically modified neural stem cells for a local and sustained delivery of neuroprotective factors to the dystrophic mouse retina. Stem Cells Transl Med 2013; 2:1001-10. [PMID: 24167317 DOI: 10.5966/sctm.2013-0013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
A continuous intraocular delivery of neurotrophic factors (NFs) is being explored as a strategy to rescue photoreceptor cells and visual functions in degenerative retinal disorders that are currently untreatable. To establish a cell-based intraocular delivery system for a sustained administration of NFs to the dystrophic mouse retina, we used a polycistronic lentiviral vector to genetically modify adherently cultivated murine neural stem (NS) cells. The vector concurrently encoded a gene of interest, a reporter gene, and a resistance gene and thus facilitated the selection, cloning, and in vivo tracking of the modified cells. To evaluate whether modified NS cells permit delivery of functionally relevant quantities of NFs to the dystrophic mouse retina, we expressed a secretable variant of ciliary neurotrophic factor (CNTF) in NS cells and grafted the cells into the vitreous space of Pde6b(rd1) and Pde6b(rd10) mice, two animal models of retinitis pigmentosa. In both mouse lines, grafted cells attached to the retina and lens, where they differentiated into astrocytes and some neurons. Adverse effects of the transplanted cells on the morphology of host retinas were not observed. Importantly, the CNTF-secreting NS cells significantly attenuated photoreceptor degeneration in both mutant mouse lines. The neuroprotective effect was significantly more pronounced when clonally derived NS cell lines selected for high expression levels of CNTF were grafted into Pde6b(rd1) mice. Intravitreal transplantations of modified NS cells may thus represent a useful method for preclinical studies aimed at evaluating the therapeutic potential of a cell-based intraocular delivery of NFs in mouse models of photoreceptor degeneration.
Collapse
|
41
|
Colella P, Auricchio A. Gene therapy of inherited retinopathies: a long and successful road from viral vectors to patients. Hum Gene Ther 2013; 23:796-807. [PMID: 22734691 DOI: 10.1089/hum.2012.123] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Inherited retinopathies (IRs) are common and untreatable blinding conditions inherited mostly as monogenic due to mutations in genes expressed in retinal photoreceptors (PRs) and in retinal pigment epithelium (RPE). Over the last two decades, the retina has emerged as one of the most favorable target tissues for gene therapy given its small size and its enclosed and immune-privileged environment. Different types of viral vectors have been developed, especially those based on the adeno-associated virus (AAV), which efficiently deliver therapeutic genes to PRs or RPE upon subretinal injections. Dozens of successful proofs of concept of the efficacy of gene therapy for recessive and dominant IRs have been generated in small and large models that have paved the way to the first clinical trials using AAV in patients with Leber congenital amaurosis, a severe form of childhood blindness. The results from these initial trials suggest that retinal gene therapy with AAV is safe in humans, that vision can be improved in patients that have suffered from severe impairment of visual function, in some cases for decades, and that readministration of AAV to the subretinal space is feasible, effective, and safe. However, none of the trials could match the levels of efficacy of gene therapy observed in a dog model of the disease, suggesting that there is room for improvement. In conclusion, these results bode well for further testing of AAV-mediated retinal gene therapy in patients with other monogenic and complex forms of blindness.
Collapse
|
42
|
Transient photoreceptor deconstruction by CNTF enhances rAAV-mediated cone functional rescue in late stage CNGB3-achromatopsia. Mol Ther 2013; 21:1131-41. [PMID: 23568263 DOI: 10.1038/mt.2013.50] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Achromatopsia is a genetic disorder of cones, and one of the most common forms is a channelopathy caused by mutations in the β-subunit, CNGB3, of the cone cyclic nucleotide-gated (CNG) channel. Recombinant adeno-associated virus of serotype 5 (rAAV5)-mediated gene transfer of human CNGB3 cDNA to mutant dog cones results in functional and structural rescue in dogs <0.5 years of age, but treatment is minimally effective in dogs >1 year. We now test a new therapeutic concept by combining gene therapy with the administration of ciliary neurotrophic factor (CNTF). Intravitreal CNTF causes transient dedifferentiation of photoreceptors, a process called deconstruction, whereby visual cells become immature with short outer segments, and decreased retinal function and gene expression that subsequently return to normal. Cone function was successfully rescued in all mutant dogs treated between 14 and 42 months of age with this strategy. CNTF-mediated deconstruction and regeneration of the photoreceptor outer segments prepares the mutant cones optimally for gene augmentation therapy.
Collapse
|
43
|
Reprogramming of adult rod photoreceptors prevents retinal degeneration. Proc Natl Acad Sci U S A 2013; 110:1732-7. [PMID: 23319618 DOI: 10.1073/pnas.1214387110] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
A prime goal of regenerative medicine is to direct cell fates in a therapeutically useful manner. Retinitis pigmentosa is one of the most common degenerative diseases of the eye and is associated with early rod photoreceptor death followed by secondary cone degeneration. We hypothesized that converting adult rods into cones, via knockdown of the rod photoreceptor determinant Nrl, could make the cells resistant to the effects of mutations in rod-specific genes, thereby preventing secondary cone loss. To test this idea, we engineered a tamoxifen-inducible allele of Nrl to acutely inactivate the gene in adult rods. This manipulation resulted in reprogramming of rods into cells with a variety of cone-like molecular, histologic, and functional properties. Moreover, reprogramming of adult rods achieved cellular and functional rescue of retinal degeneration in a mouse model of retinitis pigmentosa. These findings suggest that elimination of Nrl in adult rods may represent a unique therapy for retinal degeneration.
Collapse
|
44
|
|
45
|
Chucair-Elliott AJ, Elliott MH, Wang J, Moiseyev GP, Ma JX, Politi LE, Rotstein NP, Akira S, Uematsu S, Ash JD. Leukemia inhibitory factor coordinates the down-regulation of the visual cycle in the retina and retinal-pigmented epithelium. J Biol Chem 2012; 287:24092-102. [PMID: 22645143 DOI: 10.1074/jbc.m112.378240] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Leukemia inhibitory factor (LIF), an interleukin-6 family neurocytokine, is up-regulated in response to different types of retinal stress and has neuroprotective activity through activation of the gp130 receptor/STAT3 pathway. We observed that LIF induces rapid, robust, and sustained activation of STAT3 in both the retina and retinal pigmented epithelium (RPE). Here, we tested whether LIF-induced STAT3 activation within the RPE can down-regulate RPE65, the central enzyme in the visual cycle that provides the 11-cis-retinal chromophore to photoreceptors in vivo. We generated conditional knock-out mice to specifically delete STAT3 or gp130 in RPE, retina, or both RPE and retina. After intravitreal injection of LIF, we analyzed the expression levels of visual cycle genes and proteins, isomerase activity of RPE65, levels of rhodopsin protein, and the rates of dark adaptation and rhodopsin regeneration. We found that RPE65 protein levels and isomerase activity were reduced and recovery of bleachable rhodopsin was delayed in LIF-injected eyes. In mice with functional gp130/STAT3 signaling in the retina, rhodopsin protein was also reduced by LIF. However, the LIF-induced down-regulation of RPE65 required a functional gp130/STAT3 cascade intrinsic to RPE. Our data demonstrate that a single cytokine, LIF, can simultaneously and independently affect both RPE and photoreceptors through the same signaling cascade to reduce the generation and utilization of 11-cis-retinal.
Collapse
Affiliation(s)
- Ana J Chucair-Elliott
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Stanniocalcin-1 rescued photoreceptor degeneration in two rat models of inherited retinal degeneration. Mol Ther 2012; 20:788-97. [PMID: 22294148 DOI: 10.1038/mt.2011.308] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Oxidative stress and photoreceptor apoptosis are prominent features of many forms of retinal degeneration (RD) for which there are currently no effective therapies. We previously observed that mesenchymal stem/stromal cells reduce apoptosis by being activated to secrete stanniocalcin-1 (STC-1), a multifunctional protein that reduces oxidative stress by upregulating mitochondrial uncoupling protein-2 (UCP-2). Therefore, we tested the hypothesis that intravitreal injection of STC-1 can rescue photoreceptors. We first tested STC-1 in the rhodopsin transgenic rat characterized by rapid photoreceptor loss. Intravitreal STC-1 decreased the loss of photoreceptor nuclei and transcripts and resulted in measurable retinal function when none is otherwise present in this rapid degeneration. We then tested STC-1 in the Royal College of Surgeons (RCS) rat characterized by a slower photoreceptor degeneration. Intravitreal STC-1 reduced the number of pyknotic nuclei in photoreceptors, delayed the loss of photoreceptor transcripts, and improved function of rod photoreceptors. Additionally, STC-1 upregulated UCP-2 and decreased levels of two protein adducts generated by reactive oxygen species (ROS). Microarrays from the two models demonstrated that STC-1 upregulated expression of a similar profile of genes for retinal development and function. The results suggested that intravitreal STC-1 is a promising therapy for various forms of RD including retinitis pigmentosa and atrophic age-related macular degeneration (AMD).
Collapse
|
47
|
Wen R, Tao W, Li Y, Sieving PA. CNTF and retina. Prog Retin Eye Res 2011; 31:136-51. [PMID: 22182585 DOI: 10.1016/j.preteyeres.2011.11.005] [Citation(s) in RCA: 159] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2011] [Revised: 10/29/2011] [Accepted: 11/17/2011] [Indexed: 11/15/2022]
Abstract
Ciliary neurotrophic factor (CNTF) is one of the most studied neurotrophic factors for neuroprotection of the retina. A large body of evidence demonstrates that CNTF promotes rod photoreceptor survival in almost all animal models. Recent studies indicate that CNTF also promotes cone photoreceptor survival and cone outer segment regeneration in the degenerating retina and improves cone function in dogs with congenital achromotopsia. In addition, CNTF is a neuroprotective factor and an axogenesis factor for retinal ganglion cells (RGCs). This review focuses on the effects of exogenous CNTF on photoreceptors and RGCs in the mammalian retina and the potential clinical application of CNTF for retinal degenerative diseases.
Collapse
Affiliation(s)
- Rong Wen
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, FL 33136, USA.
| | | | | | | |
Collapse
|
48
|
Koh SWM. Corneal endothelial autocrine trophic factor VIP in a mechanism-based strategy to enhance human donor cornea preservation for transplantation. Exp Eye Res 2011; 95:48-53. [PMID: 22036689 DOI: 10.1016/j.exer.2011.10.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 10/03/2011] [Accepted: 10/14/2011] [Indexed: 12/13/2022]
Abstract
Vasoactive intestinal peptide (VIP) and ciliary neurotrophic factor (CNTF) are identified as autocrines of human corneal endothelial (CE) cells working in concert to maintain the differentiated state and promote the survival of the corneal endothelium. From VIP gene knockdown study, endogenous VIP is shown to maintain the level of the differentiation marker, the adhesion molecule N-cadherin, CE cell size, shape, and retention, in situ in the human donor corneoscleral explants. Exogenous VIP protects the corneal endothelium against the killing effect of oxidative stress, in part by upholding ATP levels in CE cells dying of oxidative stress-induced injury, allowing them to die of an apoptotic death instead of an acute necrotic one. The switch from the acute necrosis to the programmed cell death (apoptosis) may have allowed the injured CE cell to be rescued by the VIP-upregulated pathways, including those of Bcl-2 and N-cadherin, and resulted in long-term CE cell survival. The endogenous VIP in CE cells is upregulated by CNTF, which is released by CE cells surviving the oxidative stress. The CNTF receptor (CNTFRα) is expressed in CE cells in human donor corneoscleral explant and gradually becomes lost during corneal storage. VIP treatment (10(-8) M, 37 °C, 30 min) prior to storage of freshly dissected human donor corneoscleral explants increases their CE cell CNTFRα level and responsiveness to CNTF in upregulating the gap junctional protein connexin-43 expression. VIP treatment of both fresh and preserved corneoscleral explants reduces CE damage in the corneoscleral explants and in the corneal buttons trephined from them. CE cell loss is a critical risk factor in corneal graft failure at any time in the life of the graft, which can be as late as 5-10 years after an initially successful transplant. A new procedure, Descemet's stripping automated endothelial keratoplasty (DSAEK), which is superior to the traditional full thickness transplantation in many aspects, nevertheless subjects the corneal endothelium to extensive mechanical forces, resulting in even more pronounced CE cell loss than the traditional technique. Whereas it is known that cells transduce mechanical stress through N-cadherin, stimulation of the N-cadherin pathway increases the anti-apoptotic protein Bcl-2 expression. Since N-cadherin and Bcl-2 in the corneal endothelium are both upregulated by VIP, we aim to strengthen the CE sheet by VIP treatments of the corneoscleral explants for full thickness traditional corneal transplantation and pre-cut corneas for DSAEK.
Collapse
Affiliation(s)
- Shay-Whey Margaret Koh
- Department of Ophthalmology & Visual Sciences, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
49
|
Li R, Wen R, Banzon T, Maminishkis A, Miller SS. CNTF mediates neurotrophic factor secretion and fluid absorption in human retinal pigment epithelium. PLoS One 2011; 6:e23148. [PMID: 21912637 PMCID: PMC3166283 DOI: 10.1371/journal.pone.0023148] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Accepted: 07/07/2011] [Indexed: 12/14/2022] Open
Abstract
Ciliary neurotrophic factor (CNTF) protects photoreceptors and regulates their phototransduction machinery, but little is known about CNTF's effects on retinal pigment epithelial (RPE) physiology. Therefore, we determined the expression and localization of CNTF receptors and the physiological consequence of their activation in primary cultures of human fetal RPE (hfRPE). Cultured hfRPE express CNTF, CT1, and OsM and their receptors, including CNTFRα, LIFRβ, gp130, and OsMRβ, all localized mainly at the apical membrane. Exogenous CNTF, CT1, or OsM induces STAT3 phosphorylation, and OsM also induces the phosphorylation of ERK1/2 (p44/42 MAP kinase). CNTF increases RPE survivability, but not rates of phagocytosis. CNTF increases secretion of NT3 to the apical bath and decreases that of VEGF, IL8, and TGFβ2. It also significantly increases fluid absorption (JV) across intact monolayers of hfRPE by activating CFTR chloride channels at the basolateral membrane. CNTF induces profound changes in RPE cell biology, biochemistry, and physiology, including the increase in cell survival, polarized secretion of cytokines/neurotrophic factors, and the increase in steady-state fluid absorption mediated by JAK/STAT3 signaling. In vivo, these changes, taken together, could serve to regulate the microenvironment around the distal retinal/RPE/Bruch's membrane complex and provide protection against neurodegenerative disease.
Collapse
Affiliation(s)
- Rong Li
- National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Rong Wen
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
| | - Tina Banzon
- National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Arvydas Maminishkis
- National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Sheldon S. Miller
- National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
50
|
Koh SWM, Gloria D, Molloy J. Corneal endothelial autocrine VIP enhances its integrity in stored human donor corneoscleral explant. Invest Ophthalmol Vis Sci 2011; 52:5632-40. [PMID: 21482640 DOI: 10.1167/iovs.10-5983] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE To demonstrate corneal endothelial (CE) integrity enhanced during eye banking by a brief treatment of human donor corneoscleral explant (explant) with CE autocrine trophic factor vasoactive intestinal peptide (VIP). METHODS Paired explants were used as control versus VIP (10 nM)-treated before storage in corneal storage medium (4°C). CE ciliary neurotrophic factor receptor (CNTFRα) and CNTF (0.83 nM) responsiveness in connexin 43 upregulation were monitored (Western blot analysis). CE damage in CNTF-modulated explants and corneal buttons from explants was quantified by analysis of panoramic and microscopic images of the alizarin red-stained corneal endothelium. CE cells scraped from the Descemet's membrane were counted. CE VIP receptor was demonstrated (Western blot analysis). RESULTS CE cells in every VIP-treated, freshly dissected explant demonstrated higher CNTFRα levels than controls (100% vs. 142% ± 15%; P = 0.014; 7 pairs stored for 4 to 25 days). Nine days after VIP treatment of previously preserved explants, CNTF responsiveness was 174% ± 23% (P = 0.023; 4 pairs) of controls. Panoramic images of explants and corneal buttons revealed that VIP treatment reduced CE damage to 75% ± 6% (P = 0.023; 4 pairs) and 71% ± 11% (P = 0.016; 9 pairs) of controls, respectively, whereas CE damage to 39% (2 pairs) and 23% ± 4% (P < 0.001; 7 pairs), respectively, was revealed in microscopic images. Twenty-one days after VIP treatment of previously preserved explants, CE cell retention was 206% ± 38% (P = 0.008; 14 pairs) of the control. CE cells from human donor corneas expressed VIP receptor VPAC1 (not VPAC2). CONCLUSIONS CE integrity during eye banking was enhanced by a brief treatment of the explant with the CE autocrine VIP.
Collapse
Affiliation(s)
- Shay-Whey M Koh
- Department of Ophthalmology and Visual Sciences, University of Maryland School of Medicine, 10 S. Pine Street, Baltimore, MD 21201, USA.
| | | | | |
Collapse
|