1
|
Yi LY, Hsieh HH, Lin ZQ, Hung KF, Sun YC. Exploring the Role of ROCK Inhibition in Corneal Edema Through Crosstalk Between Epithelial and Endothelial Cells. J Ophthalmol 2024; 2024:9381303. [PMID: 39534682 PMCID: PMC11557173 DOI: 10.1155/2024/9381303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 09/29/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
The maintenance of corneal transparency and normal vision is dependent on preservation of epithelial and endothelial cell layer homeostases. Different types of corneal injury can induce swelling and losses in transparency. Fuchs endothelial corneal dystrophy (FECD) is one type of injury that is commonly treated with rho-associated coiled-coil-containing protein kinase (ROCK) inhibitors. While their clinical benefit is apparent, certain aspects of their mechanism of action require clarification. Specifically, although topical eye drops containing ROCK inhibitors have been employed to treat corneal endothelial dysfunction-associated corneal edema, it remains unclear whether interactions between both corneal epithelial and endothelial cell contribute to mitigating clinical signs that compromise normal vision. To address this question, we first review the intricate ROCK signaling pathways and their role in modulating a variety of functions that are related to the maintenance of corneal transparency and normal vision. We also review the results of ongoing clinical trials employing current FDA-approved ROCK inhibitors, highlighting the prominent role of Y-27632 in the treatment of a variety of ocular conditions, particularly FECD, and its promising results in reversing losses in normal vision through facilitating cell proliferation and suppressing apoptosis. This review shows that the ROCK inhibitor clinical benefit is affected by their interactions between the epithelium and the endothelium. This realization makes it likely that ROCK inhibitors will be approved for use in a clinical setting to treat FECD.
Collapse
Affiliation(s)
- Lieh-Yu Yi
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Medical Education, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - Hsiu-Hui Hsieh
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Ophthalmology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - Zhi-Qian Lin
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Kai-Feng Hung
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Dentistry, School of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Chen Sun
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Ophthalmology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| |
Collapse
|
2
|
Mokady D, Charish J, Barretto-Burns P, Grisé KN, Coles BLK, Raab S, Ortin-Martinez A, Müller A, Fasching B, Jain P, Drukker M, van der Kooy D, Steger M. Small-Molecule-Directed Endogenous Regeneration of Visual Function in a Mammalian Retinal Degeneration Model. Int J Mol Sci 2024; 25:1521. [PMID: 38338800 PMCID: PMC10855388 DOI: 10.3390/ijms25031521] [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: 10/18/2023] [Revised: 12/31/2023] [Accepted: 01/20/2024] [Indexed: 02/12/2024] Open
Abstract
Degenerative retinal diseases associated with photoreceptor loss are a leading cause of visual impairment worldwide, with limited treatment options. Phenotypic profiling coupled with medicinal chemistry were used to develop a small molecule with proliferative effects on retinal stem/progenitor cells, as assessed in vitro in a neurosphere assay and in vivo by measuring Msx1-positive ciliary body cell proliferation. The compound was identified as having kinase inhibitory activity and was subjected to cellular pathway analysis in non-retinal human primary cell systems. When tested in a disease-relevant murine model of adult retinal degeneration (MNU-induced retinal degeneration), we observed that four repeat intravitreal injections of the compound improved the thickness of the outer nuclear layer along with the regeneration of the visual function, as measured with ERG, visual acuity, and contrast sensitivity tests. This serves as a proof of concept for the use of a small molecule to promote endogenous regeneration in the eye.
Collapse
Affiliation(s)
- Daphna Mokady
- Endogena Therapeutics, Inc., 661 University Ave, Toronto, ON M5G 0B7, Canada (P.B.-B.)
| | - Jason Charish
- Endogena Therapeutics, Inc., 661 University Ave, Toronto, ON M5G 0B7, Canada (P.B.-B.)
| | | | - Kenneth N. Grisé
- Department of Molecular Genetics, University of Toronto, Donnelly Centre Rm 1110, 160 College Street, Toronto, ON M5S 3E1, Canada
| | - Brenda L. K. Coles
- Department of Molecular Genetics, University of Toronto, Donnelly Centre Rm 1110, 160 College Street, Toronto, ON M5S 3E1, Canada
| | - Susanne Raab
- Endogena Therapeutics, AG, Binzmuehlestrasse 170 d, CH-8050 Zuerich, Switzerland
| | - Arturo Ortin-Martinez
- Endogena Therapeutics, Inc., 661 University Ave, Toronto, ON M5G 0B7, Canada (P.B.-B.)
| | - Alex Müller
- Endogena Therapeutics, AG, Binzmuehlestrasse 170 d, CH-8050 Zuerich, Switzerland
| | - Bernhard Fasching
- Endogena Therapeutics, AG, Binzmuehlestrasse 170 d, CH-8050 Zuerich, Switzerland
| | - Payal Jain
- Endogena Therapeutics, Inc., 661 University Ave, Toronto, ON M5G 0B7, Canada (P.B.-B.)
| | - Micha Drukker
- Endogena Therapeutics, AG, Binzmuehlestrasse 170 d, CH-8050 Zuerich, Switzerland
| | - Derek van der Kooy
- Department of Molecular Genetics, University of Toronto, Donnelly Centre Rm 1110, 160 College Street, Toronto, ON M5S 3E1, Canada
| | - Matthias Steger
- Endogena Therapeutics, AG, Binzmuehlestrasse 170 d, CH-8050 Zuerich, Switzerland
| |
Collapse
|
3
|
Völkner M, Wagner F, Kurth T, Sykes AM, Del Toro Runzer C, Ebner LJA, Kavak C, Alexaki VI, Cimalla P, Mehner M, Koch E, Karl MO. Modeling inducible neuropathologies of the retina with differential phenotypes in organoids. Front Cell Neurosci 2023; 17:1106287. [PMID: 37213216 PMCID: PMC10196395 DOI: 10.3389/fncel.2023.1106287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 04/06/2023] [Indexed: 05/23/2023] Open
Abstract
Neurodegenerative diseases remain incompletely understood and therapies are needed. Stem cell-derived organoid models facilitate fundamental and translational medicine research. However, to which extent differential neuronal and glial pathologic processes can be reproduced in current systems is still unclear. Here, we tested 16 different chemical, physical, and cell functional manipulations in mouse retina organoids to further explore this. Some of the treatments induce differential phenotypes, indicating that organoids are competent to reproduce distinct pathologic processes. Notably, mouse retina organoids even reproduce a complex pathology phenotype with combined photoreceptor neurodegeneration and glial pathologies upon combined (not single) application of HBEGF and TNF, two factors previously associated with neurodegenerative diseases. Pharmacological inhibitors for MAPK signaling completely prevent photoreceptor and glial pathologies, while inhibitors for Rho/ROCK, NFkB, and CDK4 differentially affect them. In conclusion, mouse retina organoids facilitate reproduction of distinct and complex pathologies, mechanistic access, insights for further organoid optimization, and modeling of differential phenotypes for future applications in fundamental and translational medicine research.
Collapse
Affiliation(s)
- Manuela Völkner
- Technische Universität Dresden, Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
| | - Felix Wagner
- Technische Universität Dresden, Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
| | - Thomas Kurth
- Technische Universität Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Technology Platform Core Facility Electron Microscopy and Histology, Dresden, Germany
| | - Alex M. Sykes
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | | - Lynn J. A. Ebner
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
| | - Cagri Kavak
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
| | - Vasileia Ismini Alexaki
- Technische Universität Dresden, Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Dresden, Germany
| | - Peter Cimalla
- Technische Universität Dresden, Carl Gustav Carus Faculty of Medicine, Department of Anesthesiology and Intensive Care Medicine, Clinical Sensoring and Monitoring, Dresden, Germany
| | - Mirko Mehner
- Technische Universität Dresden, Carl Gustav Carus Faculty of Medicine, Department of Anesthesiology and Intensive Care Medicine, Clinical Sensoring and Monitoring, Dresden, Germany
| | - Edmund Koch
- Technische Universität Dresden, Carl Gustav Carus Faculty of Medicine, Department of Anesthesiology and Intensive Care Medicine, Clinical Sensoring and Monitoring, Dresden, Germany
| | - Mike O. Karl
- Technische Universität Dresden, Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
- *Correspondence: Mike O. Karl, ,
| |
Collapse
|
4
|
Karademir D, Todorova V, Ebner LJA, Samardzija M, Grimm C. Single-cell RNA sequencing of the retina in a model of retinitis pigmentosa reveals early responses to degeneration in rods and cones. BMC Biol 2022; 20:86. [PMID: 35413909 PMCID: PMC9006580 DOI: 10.1186/s12915-022-01280-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 03/12/2022] [Indexed: 11/18/2022] Open
Abstract
Background In inherited retinal disorders such as retinitis pigmentosa (RP), rod photoreceptor-specific mutations cause primary rod degeneration that is followed by secondary cone death and loss of high-acuity vision. Mechanistic studies of retinal degeneration are challenging because of retinal heterogeneity. Moreover, the detection of early cone responses to rod death is especially difficult due to the paucity of cones in the retina. To resolve heterogeneity in the degenerating retina and investigate events in both types of photoreceptors during primary rod degeneration, we utilized droplet-based single-cell RNA sequencing in an RP mouse model, rd10. Results Using trajectory analysis, we defined two consecutive phases of rod degeneration at P21, characterized by the early transient upregulation of Egr1 and the later induction of Cebpd. EGR1 was the transcription factor most significantly associated with the promoters of differentially regulated genes in Egr1-positive rods in silico. Silencing Egr1 affected the expression levels of two of these genes in vitro. Degenerating rods exhibited changes associated with metabolism, neuroprotection, and modifications to synapses and microtubules. Egr1 was also the most strongly upregulated transcript in cones. Its upregulation in cones accompanied potential early respiratory dysfunction and changes in signaling pathways. The expression pattern of EGR1 in the retina was dynamic during degeneration, with a transient increase of EGR1 immunoreactivity in both rods and cones during the early stages of their degenerative processes. Conclusion Our results identify early and late changes in degenerating rd10 rod photoreceptors and reveal early responses to rod degeneration in cones not expressing the disease-causing mutation, pointing to mechanisms relevant for secondary cone degeneration. In addition, our data implicate EGR1 as a potential key regulator of early degenerative events in rods and cones, providing a potential broad target for modulating photoreceptor degeneration. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-022-01280-9.
Collapse
Affiliation(s)
- Duygu Karademir
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Zurich, Switzerland. .,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.
| | - Vyara Todorova
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| | - Lynn J A Ebner
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Marijana Samardzija
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Christian Grimm
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
5
|
Luo LJ, Nguyen DD, Lai JY. Harnessing the tunable cavity of nanoceria for enhancing Y-27632-mediated alleviation of ocular hypertension. Theranostics 2021; 11:5447-5463. [PMID: 33859757 PMCID: PMC8039939 DOI: 10.7150/thno.54525] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 02/26/2021] [Indexed: 11/24/2022] Open
Abstract
Background: Y-27632 is a potent ophthalmic drug for the treatment of ocular hypertension, a globally prevalent eye disease. However, the sustained delivery of Y-27632 by a therapeutic carrier to lesion sites located in the inner segments of the eye for effectively treating the ocular disorder still remains challenging. Methods: To realize the goal, a strategy based on solvothermal-assisted deposition/infiltration in combination with surface modification is utilized to synthesize hollow mesoporous ceria nanoparticles (HMCNs) with tailorable shell thicknesses and drug release profiles. The shell thickness of HMCNs is rationally exploited for achieving sustained drug release and advanced therapeutic benefits. Results: The shell thickness can regulate release profiles of Y-27632, displaying that thick and thin (~40 nm and ~10 nm) shelled HMCNs reveal burst release characteristics (within 2 days) or limited drug loading content (~10% for the 40 nm thick). As a compromise, the HMCNs with moderate shell thickness (~20 nm) possess the most sustained drug release over a period of 10 days. In a rabbit model of glaucoma, a single instillation of the optimized Y-27632-loaded HMCNs can effectively treat glaucoma for 10 days via simultaneously repairing the defected cornea (recovery of ~93% ATP1A1 mRNA levels), restoring the reduced thickness of outer nuclear layer to normal (~64 µm), and restoring ~86% of the impaired photoreceptor cells. Conclusion: A comprehensive study on the importance of HMCN shell thickness in developing long-acting nano eye drops for the efficient management of glaucoma is proposed. The findings suggest a central role of nanobiomaterial structural engineering in developing the long-life eye drops for pharmacological treatment of intraocular diseases.
Collapse
|
6
|
Liu SD, Timur Y, Xu L, Meng WX, Sun B, Qiu DY. Inhibiting the ROCK Pathway Ameliorates Acute Lung Injury in Mice following Myocardial Ischemia/reperfusion. Immunol Invest 2021; 51:931-946. [PMID: 33655821 DOI: 10.1080/08820139.2021.1887887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
To clarify the role of Y-27632, a selective inhibitor of Rho-associated coiled-coil forming protein kinase (ROCK), in acute lung injury (ALI) induced by myocardial ischemia/reperfusion (I/R). Mice were randomized into Sham, I/R, and Y-27632 (10, 20 or 30 mg/kg) + I/R groups, and hemodynamics, infarcted area, the protein concentration, neutrophils in bronchoalveolar lavage fluid (BALF), malondialdehyde (MDA), superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) levels were assessed. Pathological changes were evaluated by hematoxylin-eosin (HE) staining; protein and gene expression were measured by Western blotting, enzyme-linked immunosorbent assay (ELISA), immunohistochemistry and quantitative real-time reverse-transcription polymerase chain reaction (qRT-PCR); and apoptosis was assessed by terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick-end labeling (TUNEL) staining. ROCK1 and ROCK2 expression was up-regulated in lung tissues of I/R mice compared to sham mice. Y-27632 decreased the protein concentration and the neutrophils in BALF in I/R mice, improved hemodynamics and reduced infarct size (IS)/area at risk (AAR) ratio. In addition, pathological changes in lung tissues of Y-27632-treated mice were mitigated, and these alterations were accompanied by decreases in MDA levels in lung tissues and increases in SOD and GSH-Px levels. Moreover, in I/R group, the number of apoptotic cells in lung tissue was higher than that in sham group, and p53, Caspase-3 and Bax expression was up-regulated; however, following treatment with Y-27632 (10, 20 and 30 mg/kg), these changes were reversed. Inhibition of ROCK pathway by Y-27632 ameliorated ALI in myocardial I/R mice by mitigating oxidative stress, inflammation and cell apoptosis.
Collapse
Affiliation(s)
- Shang-Dian Liu
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yagudin Timur
- Department of Pharmacology, Harbin Medical University, Harbin, China.,Department of Pharmacology, Central Laboratory of Scientific Research, Bashkir State Medical University, Ufa, Russian Federation
| | - Lei Xu
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Wei-Xin Meng
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Bo Sun
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Dong-Yun Qiu
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| |
Collapse
|
7
|
Tan J, Liu G, Lan C, Pang IH, Luo X, Wu S, Fan N, Zhang J, Wang N, Liu X. Lentiviral vector-mediated expression of C3 transferase attenuates retinal ischemia and reperfusion injury in rats. Life Sci 2021; 272:119269. [PMID: 33631175 DOI: 10.1016/j.lfs.2021.119269] [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: 11/27/2020] [Revised: 02/17/2021] [Accepted: 02/17/2021] [Indexed: 10/22/2022]
Abstract
AIMS Our previous study showed that intravitreal delivery of self-complementary AAV2 (scAAV2)-mediated exoenzyme C3 transferase (C3) can attenuate retinal ischemia/reperfusion (I/R) injury. The current study investigated the neuroprotective effects of lentivirus (LV)-mediated C3 transgene expression on rat retinal I/R injury. MAIN METHODS The LV encoding C3 and green fluorescent protein (GFP) together (LV-C3-GFP) or GFP only (LV-GFP) was intravitreally injected to SPRAGUE-DAWLEY rats. On day 5 post-intravitreal injection, eyes were evaluated by slit-lamp examination. The GFP expression on retina was confirmed by in vivo and ex vivo assessments. RhoA GTPase expression in retina was examined by western blot. Retinal I/R injury was generated by transiently increasing intraocular pressure (110 mmHg, 90 min). Eyes were then enucleated, and retinas processed for morphological analysis and TdT-dUTP terminal nick-end labeling (TUNEL) assay. KEY FINDINGS No obvious inflammatory reactions or surgical complications were observed after intravitreal injection of LV vectors. There was a significant decrease of total RhoA GTPase level in the retina treated with LV-C3-GFP. Compared to the blank control group, LV-C3-GFP and LV-GFP did not affect the retinal thickness, cell density in ganglion cell layer (GCL), or numbers of apoptotic cells in retinal flat-mounts. In the LV-GFP-treated retinas, I/R decreased the retinal thickness and GCL cell density and increased apoptotic retinal cell numbers. LV-C3-GFP significantly protected against all these degenerative effects of I/R. SIGNIFICANCE This study indicated that LV-mediated C3 transgene expression exhibits neuroprotective effects on the retinal I/R injury and holds potential as a novel neuroprotective approach targeting certain retinopathies.
Collapse
Affiliation(s)
- Junkai Tan
- Xiamen Eye Center, Xiamen University, Xiamen 361006, China
| | - Guo Liu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Chunlin Lan
- The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Iok-Hou Pang
- Department of Pharmaceutical Sciences and North Texas Eye Research Institute, University of North Texas Health Sciences Center, Fort Worth, TX 76107, United States
| | - Xiaolin Luo
- Department of Ophthalmology, the 2nd Clinical Medical College, Jinan University, Shenzhen 518020, China
| | - Shen Wu
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100005, China
| | - Ning Fan
- Shenzhen Key Laboratory of Ophthalmology, Shenzhen Eye Hospital, School of Optometry, Shenzhen University, Shenzhen 518000, China
| | - Jingxue Zhang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100005, China
| | - Ningli Wang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100005, China
| | - Xuyang Liu
- Xiamen Eye Center, Xiamen University, Xiamen 361006, China; Department of Ophthalmology, the 2nd Clinical Medical College, Jinan University, Shenzhen 518020, China.
| |
Collapse
|
8
|
Newton F, Megaw R. Mechanisms of Photoreceptor Death in Retinitis Pigmentosa. Genes (Basel) 2020; 11:genes11101120. [PMID: 32987769 PMCID: PMC7598671 DOI: 10.3390/genes11101120] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/15/2020] [Accepted: 09/22/2020] [Indexed: 02/08/2023] Open
Abstract
Retinitis pigmentosa (RP) is the most common cause of inherited blindness and is characterised by the progressive loss of retinal photoreceptors. However, RP is a highly heterogeneous disease and, while much progress has been made in developing gene replacement and gene editing treatments for RP, it is also necessary to develop treatments that are applicable to all causative mutations. Further understanding of the mechanisms leading to photoreceptor death is essential for the development of these treatments. Recent work has therefore focused on the role of apoptotic and non-apoptotic cell death pathways in RP and the various mechanisms that trigger these pathways in degenerating photoreceptors. In particular, several recent studies have begun to elucidate the role of microglia and innate immune response in the progression of RP. Here, we discuss some of the recent progress in understanding mechanisms of rod and cone photoreceptor death in RP and summarise recent clinical trials targeting these pathways.
Collapse
Affiliation(s)
- Fay Newton
- MRC Human Genetics Unit, University of Edinburgh, South Bridge, Edinburgh EH8 9YL, UK;
- Correspondence:
| | - Roly Megaw
- MRC Human Genetics Unit, University of Edinburgh, South Bridge, Edinburgh EH8 9YL, UK;
- Princess Alexandra Eye Pavilion, NHS Lothian, Edinburgh EH3 9HA, UK
| |
Collapse
|
9
|
Moura-Coelho N, Tavares Ferreira J, Bruxelas CP, Dutra-Medeiros M, Cunha JP, Pinto Proença R. Rho kinase inhibitors-a review on the physiology and clinical use in Ophthalmology. Graefes Arch Clin Exp Ophthalmol 2019; 257:1101-1117. [PMID: 30843105 DOI: 10.1007/s00417-019-04283-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 02/11/2019] [Accepted: 02/25/2019] [Indexed: 02/07/2023] Open
Abstract
The Rho kinase (ROCK) signaling pathway is involved in several cellular events that include cell proliferation and cytoskeleton modulation leading to cell adhesion. The ROCK pathway in the human eye has been hypothesized to play important roles in corneal endothelial cell physiology and pathologic states. In addition, ROCK signaling has been identified as an important regulator of trabecular meshwork (TM) outflow, which is altered in glaucomatous eyes. These roles in corneal and glaucomatous disease states have led to the growing interest in the development of drugs selectively targeting this pathway (ROCK inhibitors). The authors provide a review of the literature on the pathobiology of the ROCK signaling in corneal endothelial disease, glaucoma, and vitreoretinal disease, as well as the clinical usefulness of ROCK inhibitors in Ophthalmology.
Collapse
Affiliation(s)
- Nuno Moura-Coelho
- Ophthalmology Department, Centro Hospitalar Universitário Lisboa Central (CHULC), Alameda Santo António Capuchos, 1169-050, Lisbon, Portugal. .,Faculty of Medical Sciences
- NOVA Medical School-Nova University of Lisbon (FCM
- NMS-UNL), Lisbon, Portugal. .,Instituto Português de Retina (IPR), Lisbon, Portugal. .,Associação Médica Olhar Bem (AMO Bem), Lisbon, Portugal.
| | - Joana Tavares Ferreira
- Ophthalmology Department, Centro Hospitalar Universitário Lisboa Central (CHULC), Alameda Santo António Capuchos, 1169-050, Lisbon, Portugal.,Faculty of Medical Sciences
- NOVA Medical School-Nova University of Lisbon (FCM
- NMS-UNL), Lisbon, Portugal.,Associação Médica Olhar Bem (AMO Bem), Lisbon, Portugal
| | - Carolina Pereira Bruxelas
- Faculty of Medical Sciences
- NOVA Medical School-Nova University of Lisbon (FCM
- NMS-UNL), Lisbon, Portugal.,Ophthalmology Department, Ocidental Lisbon Hospital Center (CHLO), Lisbon, Portugal
| | - Marco Dutra-Medeiros
- Ophthalmology Department, Centro Hospitalar Universitário Lisboa Central (CHULC), Alameda Santo António Capuchos, 1169-050, Lisbon, Portugal.,Faculty of Medical Sciences
- NOVA Medical School-Nova University of Lisbon (FCM
- NMS-UNL), Lisbon, Portugal.,Instituto Português de Retina (IPR), Lisbon, Portugal.,Associação Protectora dos Diabéticos de Portugal (APDP), Lisbon, Portugal
| | - João Paulo Cunha
- Ophthalmology Department, Centro Hospitalar Universitário Lisboa Central (CHULC), Alameda Santo António Capuchos, 1169-050, Lisbon, Portugal.,Faculty of Medical Sciences
- NOVA Medical School-Nova University of Lisbon (FCM
- NMS-UNL), Lisbon, Portugal.,Associação Médica Olhar Bem (AMO Bem), Lisbon, Portugal
| | - Rita Pinto Proença
- Ophthalmology Department, Centro Hospitalar Universitário Lisboa Central (CHULC), Alameda Santo António Capuchos, 1169-050, Lisbon, Portugal.,Associação Médica Olhar Bem (AMO Bem), Lisbon, Portugal.,Faculdade de Medicina de Lisboa-Universidade de Lisboa (FML-UL), Lisbon, Portugal
| |
Collapse
|
10
|
Toms M, Burgoyne T, Tracey-White D, Richardson R, Dubis AM, Webster AR, Futter C, Moosajee M. Phagosomal and mitochondrial alterations in RPE may contribute to KCNJ13 retinopathy. Sci Rep 2019; 9:3793. [PMID: 30846767 PMCID: PMC6405871 DOI: 10.1038/s41598-019-40507-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 02/18/2019] [Indexed: 12/29/2022] Open
Abstract
Mutations in KCNJ13 are associated with two retinal disorders; Leber congenital amaurosis (LCA) and snowflake vitreoretinal degeneration (SVD). We examined the retina of kcnj13 mutant zebrafish (obelixtd15, c.502T > C p.[Phe168Leu]) to provide new insights into the pathophysiology underlying these conditions. Detailed phenotyping of obelixtd15 fish revealed a late onset retinal degeneration at 12 months. Electron microscopy of the obelixtd15 retinal pigment epithelium (RPE) uncovered reduced phagosome clearance and increased mitochondrial number and size prior any signs of retinal degeneration. Melanosome distribution was also affected in dark-adapted 12-month obelixtd15 fish. At 6 and 12 months, ATP levels were found to be reduced along with increased expression of glial fibrillary acidic protein and heat shock protein 60. Quantitative RT-PCR of polg2, fis1, opa1, sod1/2 and bcl2a from isolated retina showed expression changes consistent with altered mitochondrial activity and retinal stress. We propose that the retinal disease in this model is primarily a failure of phagosome physiology with a secondary mitochondrial dysfunction. Our findings suggest that alterations in the RPE and photoreceptor cellular organelles may contribute to KCNJ13-related retinal degeneration and provide a therapeutic target.
Collapse
Affiliation(s)
- Maria Toms
- UCL Institute of Ophthalmology, London, UK
| | | | | | | | - Adam M Dubis
- UCL Institute of Ophthalmology, London, UK.,Moorfields Eye Hospital NHS Foundation Trust, London, UK
| | - Andrew R Webster
- UCL Institute of Ophthalmology, London, UK.,Moorfields Eye Hospital NHS Foundation Trust, London, UK
| | | | - Mariya Moosajee
- UCL Institute of Ophthalmology, London, UK. .,Moorfields Eye Hospital NHS Foundation Trust, London, UK. .,Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.
| |
Collapse
|
11
|
Cell Death Pathways in Mutant Rhodopsin Rat Models Identifies Genotype-Specific Targets Controlling Retinal Degeneration. Mol Neurobiol 2018; 56:1637-1652. [PMID: 29911255 DOI: 10.1007/s12035-018-1192-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 06/08/2018] [Indexed: 12/24/2022]
Abstract
Retinitis pigmentosa (RP) is a group of inherited neurological disorders characterized by rod photoreceptor cell death, followed by secondary cone cell death leading to progressive blindness. Currently, there are no viable treatment options for RP. Due to incomplete knowledge of the molecular signaling pathways associated with RP pathogenesis, designing therapeutic strategies remains a challenge. In particular, preventing secondary cone photoreceptor cell loss is a key goal in designing potential therapies. In this study, we identified the main drivers of rod cell death and secondary cone loss in the transgenic S334ter rhodopsin rat model, tested the efficacy of specific cell death inhibitors on retinal function, and compared the effect of combining drugs to target multiple pathways in the S334ter and P23H rhodopsin rat models. The primary driver of early rod cell death in the S334ter model was a caspase-dependent process, whereas cone cell death occurred though RIP3-dependent necroptosis. In comparison, rod cell death in the P23H model was via necroptotic signaling, whereas cone cell loss occurred through inflammasome activation. Combination therapy of four drugs worked better than the individual drugs in the P23H model but not in the S334ter model. These differences imply that treatment modalities need to be tailored for each genotype. Taken together, our data demonstrate that rationally designed genotype-specific drug combinations will be an important requisite to effectively target primary rod cell loss and more importantly secondary cone survival.
Collapse
|
12
|
Nourinia R, Nakao S, Zandi S, Safi S, Hafezi-Moghadam A, Ahmadieh H. ROCK inhibitors for the treatment of ocular diseases. Br J Ophthalmol 2017; 102:bjophthalmol-2017-310378. [PMID: 28794073 DOI: 10.1136/bjophthalmol-2017-310378] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 07/10/2017] [Accepted: 07/22/2017] [Indexed: 11/03/2022]
Abstract
The Rho-kinase/ROCK (Rho-associated coiled-coil-containing protein kinase) pathway is involved in the pathogenesis of multiple ocular and systemic disorders. Recently, ROCK inhibitors have been suggested as novel treatments for various ocular diseases. Several in vitro, in vivo and clinical studies have demonstrated the safety and efficacy of ROCK inhibitors in the management of ocular disorders such as corneal epithelial and endothelial damage, glaucoma, retinal and choroidal neovascularisation, diabetic macular oedema and optic nerve disorders. In this review, these studies are explored with focus on the relevant clinical investigations.
Collapse
Affiliation(s)
- Ramin Nourinia
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shintaro Nakao
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Center for Excellence in Functional and Molecular Imaging, Brigham and Women's Hospital, Harvard University, Boston, Massachusetts, USA
| | - Souska Zandi
- Center for Excellence in Functional and Molecular Imaging, Brigham and Women's Hospital, Harvard University, Boston, Massachusetts, USA
- Swiss Eye Institute and Clinic for Vitreoretinal Diseases, Berner Augenklinik am Lindenhofspital, Bern, Switzerland
| | - Sare Safi
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Hafezi-Moghadam
- Center for Excellence in Functional and Molecular Imaging, Brigham and Women's Hospital, Harvard University, Boston, Massachusetts, USA
| | - Hamid Ahmadieh
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Townes-Anderson E, Wang J, Halász É, Sugino I, Pitler A, Whitehead I, Zarbin M. Fasudil, a Clinically Used ROCK Inhibitor, Stabilizes Rod Photoreceptor Synapses after Retinal Detachment. Transl Vis Sci Technol 2017; 6:22. [PMID: 28660097 PMCID: PMC5482187 DOI: 10.1167/tvst.6.3.22] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Accepted: 05/08/2017] [Indexed: 12/20/2022] Open
Abstract
Purpose Retinal detachment disrupts the rod-bipolar synapse in the outer plexiform layer by retraction of rod axons. We showed that breakage is due to RhoA activation whereas inhibition of Rho kinase (ROCK), using Y27632, reduces synaptic damage. We test whether the ROCK inhibitor fasudil, used for other clinical applications, can prevent synaptic injury after detachment. Methods Detachments were made in pigs by subretinal injection of balanced salt solution (BSS) or fasudil (1, 10 mM). In some animals, fasudil was injected intravitreally after BSS-induced detachment. After 2 to 4 hours, retinae were fixed for immunocytochemistry and confocal microscopy. Axon retraction was quantified by imaging synaptic vesicle label in the outer nuclear layer. Apoptosis was analyzed using propidium iodide staining. For biochemical analysis by Western blotting, retinal explants, detached from retinal pigmented epithelium, were cultured for 2 hours. Results Subretinal injection of fasudil (10 mM) reduced retraction of rod spherules by 51.3% compared to control detachments (n = 3 pigs, P = 0.002). Intravitreal injection of 10 mM fasudil, a more clinically feasible route of administration, also reduced retraction (28.7%, n = 5, P < 0.05). Controls had no photoreceptor degeneration at 2 hours, but by 4 hours apoptosis was evident. Fasudil 10 mM reduced pyknotic nuclei by 55.7% (n = 4, P < 0.001). Phosphorylation of cofilin and myosin light chain, downstream effectors of ROCK, was decreased with 30 μM fasudil (n = 8–10 explants, P < 0.05). Conclusions Inhibition of ROCK signaling with fasudil reduced photoreceptor degeneration and preserved the rod-bipolar synapse after retinal detachment. Translational Relevance These results support the possibility, previously tested with Y27632, that ROCK inhibition may attenuate synaptic damage in iatrogenic detachments.
Collapse
Affiliation(s)
- Ellen Townes-Anderson
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Jianfeng Wang
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Éva Halász
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Ilene Sugino
- Institute of Ophthalmology and Visual Science, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Amy Pitler
- Department of Microbiology, Biochemistry, and Medical Genetics, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Ian Whitehead
- Department of Microbiology, Biochemistry, and Medical Genetics, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Marco Zarbin
- Institute of Ophthalmology and Visual Science, Rutgers New Jersey Medical School, Newark, NJ, USA
| |
Collapse
|
14
|
Rocco ML, Balzamino BO, Esposito G, Petrella C, Aloe L, Micera A. NGF/anti-VEGF combined exposure protects RCS retinal cells and photoreceptors that underwent a local worsening of inflammation. Graefes Arch Clin Exp Ophthalmol 2016; 255:567-574. [PMID: 28013393 DOI: 10.1007/s00417-016-3567-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 11/26/2016] [Accepted: 12/06/2016] [Indexed: 11/26/2022] Open
Abstract
PURPOSE Our previous study highlighted the potential nerve growth factor (NGF) effect on damaged photoreceptors from a rat model of spontaneous Retinitis Pigmentosa (RP). Herein, we tested the combined NGF/anti-vascular endothelial growth factor (αVEGF) effect on cultured retinal cells isolated from Royal College of Surgeons (RCS) rats receiving an intravitreal VEGF injection (iv-VEGF) to exacerbate retinal inflammation/neovascularization. METHODS RCS (n = 75) rats were equally grouped as untreated (n = 25), iv-saline (single saline intravitreal injection; n = 25) and iv-VEGF (single VEGF intravitreal injection; n = 25). Morphological and biochemical analysis or in vitro stimulations with the biomolecular investigation were carried out on explanted retinas. Isolated retinal cells were treated with NGF and αVEGF, either alone or in combination, for 6 days and cells were harvested for morphological and biomolecular analyses. RESULTS Infiltrating inflammatory cells were detected in iv-VEGF exposed RCS retinas, indicative of exacerbated inflammation and neovascularization. In cell cultures, NGF/αVEGF significantly increased retinal cell survival as well as rhodopsin expression and neurite outgrowth in photoreceptors. Particularly, NGF/αVEGF upregulated Bcl-2 mRNA, downregulated Bax mRNA, upregulated trkANGFR mRNA and finally upregulated both NGF mRNA and protein. CONCLUSIONS These data confirm and extend our previous findings on NGF-photoreceptor crosstalk, highlighting that the NGF/αVEGF combination might be an interesting approach for improving neuroprotection of RCS retinal cells and likewise photoreceptors in the presence of neovascularization. Further studies are required to translate this in vitro approach into clinical practice.
Collapse
Affiliation(s)
| | - Bijorn Omar Balzamino
- Research Laboratories in Ophthalmology, IRCCS-G.B. Bietti Foundation, Via Santo Stefano Rotondo, 6 I-00184, Rome, Italy
| | - Graziana Esposito
- Research Laboratories in Ophthalmology, IRCCS-G.B. Bietti Foundation, Via Santo Stefano Rotondo, 6 I-00184, Rome, Italy
| | - Carla Petrella
- Institute of Cell Biology and Neurobiology, CNR, Rome, Italy
| | - Luigi Aloe
- Institute of Cell Biology and Neurobiology, CNR, Rome, Italy
| | - Alessandra Micera
- Research Laboratories in Ophthalmology, IRCCS-G.B. Bietti Foundation, Via Santo Stefano Rotondo, 6 I-00184, Rome, Italy.
| |
Collapse
|
15
|
Croze RH, Thi WJ, Clegg DO. ROCK Inhibition Promotes Attachment, Proliferation, and Wound Closure in Human Embryonic Stem Cell-Derived Retinal Pigmented Epithelium. Transl Vis Sci Technol 2016; 5:7. [PMID: 27917311 PMCID: PMC5132148 DOI: 10.1167/tvst.5.6.7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 10/10/2016] [Indexed: 12/27/2022] Open
Abstract
Purpose Nonexudative (dry) age-related macular degeneration (AMD), a leading cause of blindness in the elderly, is associated with the loss of retinal pigmented epithelium (RPE) cells and the development of geographic atrophy, which are areas devoid of RPE cells and photoreceptors. One possible treatment option would be to stimulate RPE attachment and proliferation to replace dying/dysfunctional RPE and bring about wound repair. Clinical trials are underway testing injections of RPE cells derived from pluripotent stem cells to determine their safety and efficacy in treating AMD. However, the factors regulating RPE responses to AMD-associated lesions are not well understood. Here, we use cell culture to investigate the role of RhoA coiled coil kinases (ROCKs) in human embryonic stem cell–derived RPE (hESC-RPE) attachment, proliferation, and wound closure. Methods H9 hESC were spontaneously differentiated into RPE cells. hESC-RPE cells were treated with a pan ROCK1/2 or a ROCK2 only inhibitor; attachment, and proliferation and cell size within an in vitro scratch assay were examined. Results Pharmacological inhibition of ROCKs promoted hESC-RPE attachment and proliferation, and increased the rate of closure of in vitro wounds. ROCK inhibition decreased phosphorylation of cofilin and myosin light chain, suggesting that regulation of the cytoskeleton underlies the mechanism of action of ROCK inhibition. Conclusions ROCK inhibition promotes attachment, proliferation, and wound closure in H9 hESC-RPE cells. ROCK isoforms may have different roles in wound healing. Translational Relevance Modulation of the ROCK-cytoskeletal axis has potential in stimulating wound repair in transplanted RPE cells and attachment in cellular therapies.
Collapse
Affiliation(s)
- Roxanne H Croze
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, Department of Molecular, Cellular & Developmental Biology, University of California, Santa Barbara, CA, USA
| | - William J Thi
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, Department of Molecular, Cellular & Developmental Biology, University of California, Santa Barbara, CA, USA
| | - Dennis O Clegg
- Center for Stem Cell Biology and Engineering, Neuroscience Research Institute, Department of Molecular, Cellular & Developmental Biology, University of California, Santa Barbara, CA, USA
| |
Collapse
|