1
|
Liu Y, Dong X, Wu B, Cheng Z, Zhang J, Wang J. Promising Pharmacological Interventions for Posterior Capsule Opacification: A Review. GLOBAL CHALLENGES (HOBOKEN, NJ) 2024; 8:2400181. [PMID: 39679290 PMCID: PMC11637782 DOI: 10.1002/gch2.202400181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/18/2024] [Indexed: 12/17/2024]
Abstract
Phacoemulsification combined with intraocular lens implantation is the primary treatment for cataract. Although this treatment strategy benefits patients with cataracts, posterior capsule opacification (PCO) remains a common complication that impairs vision and affects treatment outcomes. The pathogenesis of PCO is associated with the proliferation, migration, and fibrogenesis activity of residual lens epithelial cells, with epithelial-mesenchymal transition (EMT) serving as a key mechanism underlying the condition. Transforming growth factor-beta 2 (TGF-β2) is a major promotor of EMT, thereby driving PCO development. Most studies have shown that drugs and miRNAs mitigate EMT by inhibiting, clearing, or eliminating LECs. In addition, targeting EMT-related signaling pathways in TGF-β2-stimulated LECs has garnered attention as a research focus. This review highlights potential treatments for PCO and details the mechanisms by which drugs and miRNAs counter EMT.
Collapse
Affiliation(s)
- Yuxuan Liu
- AIER Cataract InstituteShenyangLiaoning Province110000China
- Shenyang Aier Ophthalmology Institute of Precision MedicineShenyangLiaoning Province110000China
- Liaoning Aier Eye HospitalShenyangLiaoning Province110000China
| | - Xiaoming Dong
- AIER Cataract InstituteShenyangLiaoning Province110000China
- Shenyang Aier Ophthalmology Institute of Precision MedicineShenyangLiaoning Province110000China
- Liaoning Aier Eye HospitalShenyangLiaoning Province110000China
| | - Bin Wu
- AIER Cataract InstituteShenyangLiaoning Province110000China
- Shenyang Aier Ophthalmology Institute of Precision MedicineShenyangLiaoning Province110000China
- Shenyang Aier Excellent Eye HospitalShenyangLiaoning Province110000China
| | - Zhigang Cheng
- AIER Cataract InstituteShenyangLiaoning Province110000China
- Shenyang Aier Ophthalmology Institute of Precision MedicineShenyangLiaoning Province110000China
- Chaoyang Aier Eye HospitalChaoyangLiaoning Province122000China
| | - Jinsong Zhang
- AIER Cataract InstituteShenyangLiaoning Province110000China
- Shenyang Aier Ophthalmology Institute of Precision MedicineShenyangLiaoning Province110000China
- Liaoning Aier Eye HospitalShenyangLiaoning Province110000China
- Shenyang Aier Excellent Eye HospitalShenyangLiaoning Province110000China
| | - Jing Wang
- AIER Cataract InstituteShenyangLiaoning Province110000China
- Shenyang Aier Ophthalmology Institute of Precision MedicineShenyangLiaoning Province110000China
- Liaoning Aier Eye HospitalShenyangLiaoning Province110000China
- Shenyang Aier Excellent Eye HospitalShenyangLiaoning Province110000China
- Aier Academy of OphthalmologyCentral South UniversityNo. 188, Furong South Road, Tianxin DistrictChangshaHunan410004P. R. China
| |
Collapse
|
2
|
Nakazawa Y, Kuno Y, Shimada H, Nagai N, Hiramatsu N, Takeda S, Yamamoto N, Funakoshi-Tago M, Sasaki H. High ambient temperature may induce presbyopia via TRPV1 activation. Med Mol Morphol 2024; 57:268-276. [PMID: 38980406 DOI: 10.1007/s00795-024-00391-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 06/12/2024] [Indexed: 07/10/2024]
Abstract
The prevalence of presbyopia and nuclear cataracts (NUC) is reported to be higher in tropical areas than that in other regions, suggesting a potential influence of high temperatures on lens health. Transient receptor potential vanilloid (TRPV) channels play a crucial role in detecting ambient temperatures across various species, with TRPV1 and TRPV4 expressed in lens epithelial cells. In this study, we investigated whether ambient temperatures affect TRPV1 and TRPV4 activity in the lens, potentially contributing to the development of presbyopia and NUC. We conducted experiments using cultured human lens epithelial cell lines under different temperature conditions. Our results revealed that the mitogen-activated protein kinase (MEK)/extracellular signal-regulated kinase (ERK) and p38 pathways, downstream molecules of TRPV1, were activated, while Src family kinase, a downstream molecule of TRPV4, was inhibited at 37.5 °C culture compared to 35.0 °C. Confocal microscope images demonstrated higher expression of TRPV1 in 3D-structured cells under high-temperature culture conditions. Additionally, in organ culture lenses, higher elasticity was observed at elevated temperatures compared to that at lower temperatures. These results suggest that high ambient temperatures may induce lens sclerosis via TRPV1 activation, potentially contributing to the development of presbyopia and NUC.
Collapse
Affiliation(s)
- Yosuke Nakazawa
- Faculty of Pharmacy, Keio University, 1-5-30, Shibako-en, Minato-ku, Tokyo, 105-8512, Japan.
| | - Yumika Kuno
- Faculty of Pharmacy, Keio University, 1-5-30, Shibako-en, Minato-ku, Tokyo, 105-8512, Japan
| | - Hibiki Shimada
- Faculty of Pharmacy, Keio University, 1-5-30, Shibako-en, Minato-ku, Tokyo, 105-8512, Japan
| | - Noriaki Nagai
- Faculty of Pharmacy, Kindai University, Osaka, Japan
| | - Noriko Hiramatsu
- Research Promotion and Support Headquarters, Fujita Health University, Toyoake, Japan
| | - Shun Takeda
- Department of Ophthalmology, Kanazawa Medical University, 1-1 Daigaku Uchinada-machi, Kahoku-gun, Ishikawa, 920-0293, Japan
| | - Naoki Yamamoto
- Research Promotion and Support Headquarters, Fujita Health University, Toyoake, Japan
| | - Megumi Funakoshi-Tago
- Faculty of Pharmacy, Keio University, 1-5-30, Shibako-en, Minato-ku, Tokyo, 105-8512, Japan
| | - Hiroshi Sasaki
- Department of Ophthalmology, Kanazawa Medical University, 1-1 Daigaku Uchinada-machi, Kahoku-gun, Ishikawa, 920-0293, Japan.
| |
Collapse
|
3
|
Zhang X, Wang F, Su Y. TRPV: An emerging target in glaucoma and optic nerve damage. Exp Eye Res 2024; 239:109784. [PMID: 38199261 DOI: 10.1016/j.exer.2024.109784] [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: 08/23/2023] [Revised: 11/30/2023] [Accepted: 01/06/2024] [Indexed: 01/12/2024]
Abstract
Transient receptor potential vanilloid (TRPV) channels are members of the TRP channel superfamily, which are ion channels that sense mechanical and osmotic stimuli and participate in Ca2+ signalling across the cell membrane. TRPV channels play important roles in maintaining the normal functions of an organism, and defects or abnormalities in TRPV channel function cause a range of diseases, including cardiovascular, neurological and urological disorders. Glaucoma is a group of chronic progressive optic nerve diseases with pathological changes that can occur in the tissues of the anterior and posterior segments of the eye, including the ciliary body, trabecular meshwork, Schlemm's canal, and retina. TRPV channels are expressed in these tissues and play various roles in glaucoma. In this article, we review various aspects of the pathogenesis of glaucoma, the structure and function of TRPV channels, the relationship between TRPV channels and systemic diseases, and the relationship between TRPV channels and ocular diseases, especially glaucoma, and we suggest future research directions. This information will help to further our understanding of TRPV channels and provide new ideas and targets for the treatment of glaucoma and optic nerve damage.
Collapse
Affiliation(s)
- Xiaotong Zhang
- Department of Ophthalmology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Feng Wang
- Department of Ophthalmology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China.
| | - Ying Su
- Eye Hospital, The First Affiliated Hospital, Harbin Medical University, Harbin, China.
| |
Collapse
|
4
|
Doki Y, Nakazawa Y, Sukegawa M, Petrova RS, Ishida Y, Endo S, Nagai N, Yamamoto N, Funakoshi-Tago M, Donaldson PJ. Piezo1 channel causes lens sclerosis via transglutaminase 2 activation. Exp Eye Res 2023; 237:109719. [PMID: 37951336 DOI: 10.1016/j.exer.2023.109719] [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: 09/01/2023] [Revised: 10/31/2023] [Accepted: 11/06/2023] [Indexed: 11/13/2023]
Abstract
Presbyopia is caused by age-related lenticular hardening, resulting in near vision loss, and it occurs in almost every individual aged ≥50 years. The lens experiences mechanical pressure during for focal adjustment to change its thickness. As lenticular stiffening results in incomplete thickness changes, near vision is reduced, which is known as presbyopia. Piezo1 is a mechanosensitive channel that constantly senses pressure changes during the regulation of visual acuity, and changes in Piezo1 channel activity may contribute to presbyopia. However, no studies have reported on Piezo1 activation or the onset of presbyopia. To elucidate the relevance of Piezo1 activation and cross-linking in the development of presbyopia, we analysed the function of Piezo1 in the lens. The addition of Yoda1, a Piezo1 activator, induced an increase in transglutaminase 2 (TGM2) mRNA expression and activity through the extra-cellular signal-regulated kinase (ERK) 1/2 and c-Jun-NH2-terminal kinase1/2 pathways. In ex vivo lenses, Yoda1 treatment induced γ-crystallin cross-linking via TMG2 activation. Furthermore, Yoda1 eye-drops in mice led to lenticular hardening via TGM2 induction and activation in vivo, suggesting that Yoda1-treated animals could serve as a model for presbyopia. Our findings indicate that this presbyopia-animal model could be useful for screening drugs for lens-stiffening inhibition.
Collapse
Affiliation(s)
- Yuri Doki
- Faculty of Pharmacy, Keio University, Tokyo, Japan
| | | | | | - Rosica S Petrova
- Department of Physiology, School of Medical Sciences, New Zealand Eye Centre, University of Auckland, New Zealand
| | - Yuki Ishida
- R&D Division, Hayashibara Co., Ltd, Okayama, Japan
| | - Shin Endo
- R&D Division, Hayashibara Co., Ltd, Okayama, Japan
| | - Noriaki Nagai
- Faculty of Pharmacy, Kindai University, Osaka, Japan
| | - Naoki Yamamoto
- Research Promotion and Support Headquarters, Fujita Health University, Toyoake, Japan
| | | | - Paul J Donaldson
- Department of Physiology, School of Medical Sciences, New Zealand Eye Centre, University of Auckland, New Zealand.
| |
Collapse
|
5
|
Sun Q, Li J, Ma J, Zheng Y, Ju R, Li X, Ren X, Huang L, Chen R, Tan X, Luo L. JAM-C Is Important for Lens Epithelial Cell Proliferation and Lens Fiber Maturation in Murine Lens Development. Invest Ophthalmol Vis Sci 2023; 64:15. [PMID: 38095908 PMCID: PMC10723223 DOI: 10.1167/iovs.64.15.15] [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: 09/03/2023] [Accepted: 11/16/2023] [Indexed: 12/17/2023] Open
Abstract
Purpose The underlying mechanism of congenital cataracts caused by deficiency or mutation of junctional adhesion molecule C (JAM-C) gene remains unclear. Our study aims to elucidate the abnormal developmental process in Jamc-/- lenses and reveal the genes related to lens development that JAM-C may regulate. Methods Jamc knockout (Jamc-/-) mouse embryos and pups were generated for in vivo studies. Four key developmental stages from embryonic day (E) 12.5 to postnatal day (P) 0.5 were selected for the following experiments. Hematoxylin and eosin staining was used for histological analysis. The 5-bromo-2'-deoxyuridine (BrdU) incorporation assay and TUNEL staining were performed to label lens epithelial cell (LEC) proliferation and apoptosis, respectively. Immunofluorescence and Western blot were used to analyze the markers of lens epithelium, cell cycle exit, and lens fiber differentiation. Results JAM-C was expressed throughout the process of lens development. Deletion of Jamc resulted in decreased lens size and disorganized lens fibers, which arose from E16.5 and aggravated gradually. The LECs of Jamc-/- lenses showed decreased quantity and proliferation, accompanied with reduction of key transcription factor, FOXE3. The fibers in Jamc-/- lenses were disorganized. Moreover, Jamc-deficient lens fibers showed significantly altered distribution patterns of Cx46 and Cx50. The marker of fiber homeostasis, γ-crystallin, was also decreased in the inner cortex and core fibers of Jamc-/- lenses. Conclusions Deletion of JAM-C exhibits malfunction of LEC proliferation and fiber maturation during murine lens development, which may be related to the downregulation of FOXE3 expression and abnormal localization patterns of Cx46 and Cx50.
Collapse
Affiliation(s)
- Qihang Sun
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jiani Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jingyu Ma
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yuxing Zheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Rong Ju
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xuri Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xiangrong Ren
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Lijuan Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Rongyuan Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xuhua Tan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Lixia Luo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| |
Collapse
|
6
|
Orman B, Benozzi G. Pharmacological Treatments for Presbyopia. Drugs Aging 2023; 40:105-116. [PMID: 36670320 DOI: 10.1007/s40266-022-01002-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2022] [Indexed: 01/22/2023]
Abstract
Accommodation is the change in dioptric power of the eye. It is a dynamic process that allows focusing on an object at all distances. In order to focus sharply, three physiological responses, known as the triad of accommodation, are produced by a change in pupil size, a change in shape and position of the lens, and ocular convergence. This is modulated by the autonomic nervous system, mainly the parasympathetic nervous system. Presbyopia is a refractive condition that occurs with aging, usually manifesting around 40-50 years of age, and is a result of the loss of accommodation in the eye, causing loss of visual performance when focusing on objects placed at different distances, starting with near vision. Glasses, contact lenses, surgical approaches and now pharmacological treatments are accepted methods of treating presbyopia. Pharmacological treatment is a promising new noninvasive option for treating presbyopia. Currently there are three pharmacological approaches to the treatment of presbyopia. The first one aims to produce miosis and, from a pinhole effect, increase depth of focus, and therefore improve uncorrected near visual acuity (UNVA). The second one addresses rehabilitating accommodation in a binocular way, allowing good vision at all distances. Finally, the third strategy uses lipoic acid to restore the lost elasticity of the lens. All of these pharmacological treatments are topical non-invasive eyedrops, with no serious adverse effects having been reported with any of the strategies, and require the right patient selection process to fulfill expectations and needs. The aim of this article is to provide an update on recent advances in this field.
Collapse
Affiliation(s)
- Betina Orman
- Universidad de Buenos Aires, Facultad de Odontología, Cátedra de Farmacología, Buenos Aires, Argentina.
| | - Giovanna Benozzi
- Centro de Investigación Avanzada para la Presbicia, Buenos Aires, Argentina
| |
Collapse
|
7
|
Ek-Vitorin JF, Shahidullah M, Lopez Rosales JE, Delamere NA. Patch clamp studies on TRPV4-dependent hemichannel activation in lens epithelium. Front Pharmacol 2023; 14:1101498. [PMID: 36909173 PMCID: PMC9998544 DOI: 10.3389/fphar.2023.1101498] [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: 11/17/2022] [Accepted: 02/06/2023] [Indexed: 02/26/2023] Open
Abstract
ATP release from the lens via hemichannels has been explained as a response to TRPV4 activation when the lens is subjected to osmotic swelling. To explore the apparent linkage between TRPV4 activation and connexin hemichannel opening we performed patch-clamp recordings on cultured mouse lens epithelial cells exposed to the TRPV4 agonist GSK1016790A (GSK) in the presence or absence of the TRPV4 antagonist HC067047 (HC). GSK was found to cause a fast, variable and generally large non-selective increase of whole cell membrane conductance evident as a larger membrane current (Im) over a wide voltage range. The response was prevented by HC. The GSK-induced Im increase was proportionally larger at negative voltages and coincided with fast depolarization and the simultaneous disappearance of an outward current, likely a K+ current. The presence of this outward current in control conditions appeared to be a reliable predictor of a cell's response to GSK treatment. In some studies, recordings were obtained from single cells by combining cell-attached and whole-cell patch clamp configurations. This approach revealed events with a channel conductance 180-270 pS following GSK application through the patch pipette on the cell-attached side. The findings are consistent with TRPV4-dependent opening of Cx43 hemichannels.
Collapse
Affiliation(s)
- Jose F Ek-Vitorin
- Department of Physiology, University of Arizona, Tucson, AZ, United States
| | - Mohammad Shahidullah
- Department of Physiology, University of Arizona, Tucson, AZ, United States.,Department of Ophthalmology and Vision Science, University of Arizona, Tucson, AZ, United States
| | | | - Nicholas A Delamere
- Department of Physiology, University of Arizona, Tucson, AZ, United States.,Department of Ophthalmology and Vision Science, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
8
|
Regulation of lens water content: Effects on the physiological optics of the lens. Prog Retin Eye Res 2022:101152. [DOI: 10.1016/j.preteyeres.2022.101152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/09/2022]
|
9
|
Paensuwan P, Laorob T, Ngoenkam J, Wichai U, Pongcharoen S. Nitro Dihydrocapsaicin, a Non-Pungent Capsaicin Analogue, Inhibits Cellular Senescence of Lens Epithelial Cells via Upregulation of SIRT1. Int J Mol Sci 2022; 23:13960. [PMID: 36430438 PMCID: PMC9695757 DOI: 10.3390/ijms232213960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/05/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Diabetic cataracts are a common complication that can cause blindness among patients with diabetes mellitus. A novel nitro dihydrocapsaicin (NDHC), a capsaicin analog, was constructed to have a non-pungency effect. The objective of this research was to study the effect of NDHC on human lens epithelial (HLE) cells that lost function from hyperglycemia. HLE cells were pretreated with NDHC before an exposure to high glucose (HG) conditions. The results show that NDHC promoted a deacceleration of cellular senescence in HLE cells. This inhibition of cellular senescence was characterized by a delayed cell growth and lower production of reactive oxygen species (ROS) as well as decreased SA-β-galactosidase activity. Additionally, the expression of Sirt1 protein sharply increased, while the expression of p21 and phospho-p38 proteins decreased. These findings provide evidence that NDHC could exert a pharmacologically protective effect by inhibiting the senescence program of lens cells during diabetic cataracts.
Collapse
Affiliation(s)
- Pussadee Paensuwan
- Department of Optometry, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Thanet Laorob
- Department of Chemistry, Faculty of Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Jatuporn Ngoenkam
- Department of Microbiology and Parasitology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Uthai Wichai
- Department of Chemistry, Faculty of Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Sutatip Pongcharoen
- Department of Medicine, Faculty of Medicine, Naresuan University, Phitsanulok 65000, Thailand
| |
Collapse
|
10
|
Suzuki-Kerr H, Walker KL, Han MH, Lim JC, Donaldson PJ. Hyposmotic stress causes ATP release in a discrete zone within the outer cortex of rat lens. Mol Vis 2022; 28:245-256. [PMID: 36284672 PMCID: PMC9514545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 09/02/2022] [Indexed: 12/02/2022] Open
Abstract
PURPOSE Purinergic signaling pathways activated by extracellular ATP have been implicated in the regulation of lens volume and transparency. In this study, we investigated the location of ATP release from whole rat lenses and the mechanism by which osmotic challenge alters such ATP release. METHODS Three-week-old rat lenses were cultured for 1 h in isotonic artificial aqueous humor (AAH) with no extracellular Ca2+, hypotonic AAH, or hypertonic AAH. The hypotonic AAH-treated lenses were also cultured in the absence or presence of connexin hemichannels and the pannexin channel blockers carbenoxolone, probenecid, and flufenamic acid. The ATP concentration in the AAH was determined using a Luciferin/luciferase bioluminescence assay. To visualize sites of ATP release induced by hemichannel and/or pannexin opening, the lenses were cultured in different AAH solutions, as described above, and incubated in the presence of Lucifer yellow (MW = 456 Da) and Texas red-dextran (MW = 10 kDa) for 1 h. Then the lenses were fixed, cryosectioned, and imaged using confocal microscopy to visualize areas of dye uptake from the extracellular space. RESULTS The incubation of the rat lenses in the AAH that lacked Ca2+ induced a significant increase in the extracellular ATP concentration. This was associated with an increased uptake of Lucifer yellow but not of Texas red-dextran in a discrete region of the outer cortex of the lens. Hypotonic stress caused a similar increase in ATP release and an increase in the uptake of Lucifer yellow in the outer cortex, which was significantly reduced by probenecid but not by carbenoxolone or flufenamic acid. CONCLUSIONS Our data suggest that in response to hypotonic stress, the intact rat lens is capable of releasing ATP. This seems to be mediated via the opening of pannexin channels in a specific zone of the outer cortex of the lens. Our results support the growing evidence that the lens actively regulates its volume and therefore, its optical properties, via puerinergic signaling pathways.
Collapse
Affiliation(s)
- Haruna Suzuki-Kerr
- Department of Physiology, School of Medical Sciences,,The New Zealand National Eye Centre, the University of Auckland, New Zealand
| | - Kerry L. Walker
- Department of Physiology, School of Medical Sciences,,The New Zealand National Eye Centre, the University of Auckland, New Zealand
| | | | - Julie C. Lim
- Department of Physiology, School of Medical Sciences,,The New Zealand National Eye Centre, the University of Auckland, New Zealand
| | - Paul J. Donaldson
- Department of Physiology, School of Medical Sciences,,The New Zealand National Eye Centre, the University of Auckland, New Zealand
| |
Collapse
|
11
|
Lapajne L, Rudzitis CN, Cullimore B, Ryskamp D, Lakk M, Redmon SN, Yarishkin O, Krizaj D. TRPV4: Cell type-specific activation, regulation and function in the vertebrate eye. CURRENT TOPICS IN MEMBRANES 2022; 89:189-219. [PMID: 36210149 PMCID: PMC9879314 DOI: 10.1016/bs.ctm.2022.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The architecture of the vertebrate eye is optimized for efficient delivery and transduction of photons and processing of signaling cascades downstream from phototransduction. The cornea, lens, retina, vasculature, ciliary body, ciliary muscle, iris and sclera have specialized functions in ocular protection, transparency, accommodation, fluid regulation, metabolism and inflammatory signaling, which are required to enable function of the retina-light sensitive tissue in the posterior eye that transmits visual signals to relay centers in the midbrain. This process can be profoundly impacted by non-visual stimuli such as mechanical (tension, compression, shear), thermal, nociceptive, immune and chemical stimuli, which target these eye regions to induce pain and precipitate vision loss in glaucoma, diabetic retinopathy, retinal dystrophies, retinal detachment, cataract, corneal dysfunction, ocular trauma and dry eye disease. TRPV4, a polymodal nonselective cation channel, integrate non-visual inputs with homeostatic and signaling functions of the eye. The TRPV4 gene is expressed in most if not all ocular tissues, which vary widely with respect to the mechanisms of TRPV4 channel activation, modulation, oligomerization, and participation in protein- and lipid interactions. Under- and overactivation of TRPV4 may affect intraocular pressure, maintenance of blood-retina barriers, lens accommodation, neuronal function and neuroinflammation. Because TRPV4 dysregulation precipitates many pathologies across the anterior and posterior eye, the channel could be targeted to mitigate vision loss.
Collapse
Affiliation(s)
- Luka Lapajne
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT, United States; Department of Ophthalmology, University Medical Centre, University of Ljubljana, Ljubljana, Slovenia
| | - Christopher N Rudzitis
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Brenan Cullimore
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Daniel Ryskamp
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Monika Lakk
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Sarah N Redmon
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Oleg Yarishkin
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - David Krizaj
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT, United States; Department of Neurobiology, University of Utah, Salt Lake City, UT, United States; Department of Bioengineering, University of Utah, Salt Lake City, UT, United States.
| |
Collapse
|
12
|
Peng G, Tang X, Gui Y, Yang J, Ye L, Wu L, Ding YH, Wang L. Transient receptor potential vanilloid subtype 1: A potential therapeutic target for fibrotic diseases. Front Physiol 2022; 13:951980. [PMID: 36045746 PMCID: PMC9420870 DOI: 10.3389/fphys.2022.951980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/11/2022] [Indexed: 11/23/2022] Open
Abstract
The transient receptor potential vanilloid subtype 1 (TRPV1), belonging to the TRPV channel family, is a non-selective, calcium-dependent, cation channel implicated in several pathophysiological processes. Collagen, an extracellular matrix component, can accumulate under pathological conditions and may lead to the destruction of tissue structure, organ dysfunction, and organ failure. Increasing evidence indicates that TRPV1 plays a role in the development and occurrence of fibrotic diseases, including myocardial, renal, pancreatic, and corneal fibrosis. However, the mechanism by which TRPV1 regulates fibrosis remains unclear. This review highlights the comprehensive role played by TRPV1 in regulating pro-fibrotic processes, the potential of TRPV1 as a therapeutic target in fibrotic diseases, as well as the different signaling pathways associated with TRPV1 and fibrosis.
Collapse
Affiliation(s)
- Guangxin Peng
- Zhejiang University of Technology, Hangzhou, China
- Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Xiaoling Tang
- Zhejiang University of Technology, Hangzhou, China
- Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Yang Gui
- Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Jing Yang
- Zhejiang University of Technology, Hangzhou, China
- Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Lifang Ye
- Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Liuyang Wu
- Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Ya hui Ding
- Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Lihong Wang
- Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
- *Correspondence: Lihong Wang,
| |
Collapse
|
13
|
Chen Y, Petrova RS, Qiu C, Donaldson PJ. -Intracellular hydrostatic pressure regulation in the bovine lens: a role in the regulation of lens optics? Am J Physiol Regul Integr Comp Physiol 2022; 322:R263-R279. [PMID: 35107027 DOI: 10.1152/ajpregu.00309.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The optical properties of the bovine lens have been shown to be actively maintained by an internal microcirculation system. In the mouse lens, this water transport through gap junction channels generates an intracellular hydrostatic pressure gradient, which is subjected to a dual feedback regulation that is mediated by the reciprocal modulation of the transient receptor potential vanilloid channels, TRPV1 and TRPV4. Here we test whether a similar feedback regulation of pressure exists in the bovine lens, and whether it regulates overall lens optics. Lens pressure was measured using a microelectrode/pico-injector-based pressure measurement system, and lens optics were monitored in organ cultured lenses using a laser ray tracing system. Like the mouse, the bovine lenses exhibited a similar pressure gradient (0 to 340 mmHg). Activation of TRPV1 with capsaicin caused a biphasic increase in surface pressure, while activation of TRPV4 with GSK1016790A caused a biphasic decrease in pressure. These biphasic responses were abolished if lenses were pre-incubated with either the TRPV1 inhibitor A-889425, or the TRPV4 inhibitor HC-067047. While modulation of lens pressure by TRPV1 and TRPV4 had minimal effects on lens power and overall vision quality, the changes in lens pressure did induce opposing changes to lens geometry and GRIN that effectively kept lens power constant. Hence, our results suggest that the TRPV1/4 mediated feedback control of lens hydrostatic pressure operates to ensure that any fluctuations in lens water transport, and consequently water content, do not result in changes in lens power and therefore overall vision quality.
Collapse
Affiliation(s)
- Yadi Chen
- Department of Physiology, School of Medical Sciences, New Zealand Eye Centre, University of Auckland, New Zealand
| | - Rosica S Petrova
- Department of Physiology, School of Medical Sciences, New Zealand Eye Centre, University of Auckland, New Zealand
| | - Chen Qiu
- Department of Physiology, School of Medical Sciences, New Zealand Eye Centre, University of Auckland, New Zealand
| | - Paul J Donaldson
- Department of Physiology, School of Medical Sciences, New Zealand Eye Centre, University of Auckland, New Zealand
| |
Collapse
|
14
|
Ebihara L, Acharya P, Tong JJ. Mechanical Stress Modulates Calcium-Activated-Chloride Currents in Differentiating Lens Cells. Front Physiol 2022; 13:814651. [PMID: 35173630 PMCID: PMC8842795 DOI: 10.3389/fphys.2022.814651] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 01/06/2022] [Indexed: 11/13/2022] Open
Abstract
During accommodation, the lens changes focus by altering its shape following contraction and relaxation of the ciliary muscle. At the cellular level, these changes in shape may be accompanied by fluid flow in and out of individual lens cells. We tested the hypothesis that some of this flow might be directly modulated by pressure-activated channels. In particular, we used the whole cell patch clamp technique to test whether calcium-activated-chloride channels (CaCCs) expressed in differentiating lens cells are activated by mechanical stimulation. Our results show that mechanical stress, produced by focally perfusing the lens cell at a constant rate, caused a significant increase in a chloride current that could be fully reversed by stopping perfusion. The time course of activation and recovery from activation of the flow-induced current occurred rapidly over a time frame similar to that of accommodation. The flow-induced current could be inhibited by the TMEM16A specific CaCC blocker, Ani9, suggesting that the affected current was predominantly due to TMEM16A chloride channels. The mechanism of action of mechanical stress did not appear to involve calcium influx through other mechanosensitive ion channels since removal of calcium from the bath solution failed to block the flow-induced chloride current. In conclusion, our results suggest that CaCCs in the lens can be rapidly and reversibly modulated by mechanical stress, consistent with their participation in regulation of volume in this organ.
Collapse
Affiliation(s)
- Lisa Ebihara
- Center of Proteomics and Molecular Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
- Discipline of Physiology and Biophysics, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
- *Correspondence: Lisa Ebihara,
| | - Pooja Acharya
- Center of Proteomics and Molecular Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Jun-Jie Tong
- Center of Proteomics and Molecular Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| |
Collapse
|
15
|
Delamere NA, Shahidullah M. Ion Transport Regulation by TRPV4 and TRPV1 in Lens and Ciliary Epithelium. Front Physiol 2022; 12:834916. [PMID: 35173627 PMCID: PMC8841554 DOI: 10.3389/fphys.2021.834916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 12/27/2021] [Indexed: 02/02/2023] Open
Abstract
Aside from a monolayer of epithelium at the anterior surface, the lens is formed by tightly compressed multilayers of fiber cells, most of which are highly differentiated and have a limited capacity for ion transport. Only the anterior monolayer of epithelial cells has high Na, K-ATPase activity. Because the cells are extensively coupled, the lens resembles a syncytium and sodium-potassium homeostasis of the entire structure is largely dependent on ion transport by the epithelium. Here we describe recent studies that suggest TRPV4 and TRPV1 ion channels activate signaling pathways that play an important role in matching epithelial ion transport activity with needs of the lens cell mass. A TRPV4 feedback loop senses swelling in the fiber mass and increases Na, K-ATPase activity to compensate. TRPV4 channel activation in the epithelium triggers opening of connexin hemichannels, allowing the release of ATP that stimulates purinergic receptors in the epithelium and results in the activation of Src family tyrosine kinases (SFKs) and SFK-dependent increase of Na, K-ATPase activity. A separate TRPV1 feedback loop senses shrinkage in the fiber mass and increases NKCC1 activity to compensate. TRPV1 activation causes calcium-dependent activation of a signaling cascade in the lens epithelium that involves PI3 kinase, ERK, Akt and WNK. TRPV4 and TRPV1 channels are also evident in the ciliary body where Na, K-ATPase is localized on one side of a bilayer in which two different cell types, non-pigmented and pigmented ciliary epithelium, function in a coordinated manner to secrete aqueous humor. TRPV4 and TRPV1 may have a role in maintenance of cell volume homeostasis as ions and water move through the bilayer.
Collapse
|
16
|
Regulation of the Membrane Trafficking of the Mechanosensitive Ion Channels TRPV1 and TRPV4 by Zonular Tension, Osmotic Stress and Activators in the Mouse Lens. Int J Mol Sci 2021; 22:ijms222312658. [PMID: 34884463 PMCID: PMC8657824 DOI: 10.3390/ijms222312658] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 12/30/2022] Open
Abstract
Lens water transport generates a hydrostatic pressure gradient that is regulated by a dual-feedback system that utilizes the mechanosensitive transient receptor potential vanilloid (TRPV) channels, TRPV1 and TRPV4, to sense changes in mechanical tension and extracellular osmolarity. Here, we investigate whether the modulation of TRPV1 or TRPV4 activity dynamically affects their membrane trafficking. Mouse lenses were incubated in either pilocarpine or tropicamide to alter zonular tension, exposed to osmotic stress, or the TRPV1 and TRPV4 activators capsaicin andGSK1016790A (GSK101), and the effect on the TRPV1 and TRPV4 membrane trafficking in peripheral fiber cells visualized using confocal microscopy. Decreases in zonular tension caused the removal of TRPV4 from the membrane of peripheral fiber cells. Hypotonic challenge had no effect on TRPV1, but increased the membrane localization of TRPV4. Hypertonic challenge caused the insertion of TRPV1 and the removal of TRPV4 from the membranes of peripheral fiber cells. Capsaicin caused an increase in TRPV4 membrane localization, but had no effect on TRPV1; while GSK101 decreased the membrane localization of TRPV4 and increased the membrane localization of TRPV1. These reciprocal changes in TRPV1/4 membrane localization are consistent with the channels acting as mechanosensitive transducers of a dual-feedback pathway that regulates lens water transport.
Collapse
|
17
|
Sugiyama Y, Nakazawa Y, Sakagami T, Kawata S, Nagai N, Yamamoto N, Funakoshi-Tago M, Tamura H. Capsaicin attenuates TGFβ2-induced epithelial-mesenchymal-transition in lens epithelial cells in vivo and in vitro. Exp Eye Res 2021; 213:108840. [PMID: 34798144 DOI: 10.1016/j.exer.2021.108840] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 11/04/2021] [Accepted: 11/09/2021] [Indexed: 01/09/2023]
Abstract
Posterior capsule opacification (PCO), the most common complication of cataract surgery occurring in 20-50% of patients after 2-5 years of cataract surgery, is a major problem in the aging society. The epithelial-mesenchymal transition (EMT) of lens epithelial cells after cataract surgery has been proposed as a major cause of PCO. Capsaicin, widely used as a food additive and analgesic agent, is a major pungent ingredient in red pepper. Although the effect of capsaicin on EMT has been reported in cancer cells, the biological reaction of capsaicin was unique in each cell type, and there have been no reports describing its effects on EMT earlier. In this study, we demonstrated that treatment with capsaicin inhibited TGFβ2-induced EMT in vitro lens epithelial cells and ex vivo explant lens epithelial cells. Furthermore, eye drops of capsaicin inhibited the PCO model mice in vivo. Finally, we showed that capsaicin inhibited non-canonically induced Smad2/3 activation via suppression of EGFR activation and ERK phosphorylation. Our findings indicate that capsaicin and its derivatives are good candidate compounds for preventing PCO after cataract surgery.
Collapse
Affiliation(s)
| | | | | | - Sara Kawata
- Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Noriaki Nagai
- Faculty of Pharmacy, Kindai University, Osaka, Japan
| | - Naoki Yamamoto
- Research Promotion and Support Headquarters, Fujita Health University, Japan
| | | | | |
Collapse
|
18
|
Koç Ş. A possible follow-up method for diabetic heart failure patients. Int J Clin Pract 2021; 75:e14794. [PMID: 34482595 DOI: 10.1111/ijcp.14794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 09/02/2021] [Indexed: 11/30/2022] Open
Abstract
INTRODUCTION Plasma osmolarity is maintained through various mechanisms. The osmolarity of the aqueous humor around the crystalline lens is correlated with plasma osmolarity. A vacuole can be formed in the lens upon changes in osmolarity. The sodium-glucose cotransporter 2 inhibitors (SGLT2i) are new in the treatment of heart failure. They can cause osmotic diuresis but do not affect plasma osmolarity. OBJECTIVE It is unclear if the presence or absence of lens vacuole changes can monitor diabetic heart failure and SGLT2i treatment efficacy. METHODS Web of Science, PubMed and Scopus databases were searched for relevant articles about osmolarity, diabetes, transient receptor potential vanilloid channel, diabetic heart failure, lens vacuoles up to May 2021. MAIN MESSAGE The effect of SGLT2i on osmosis underlies its benefit to heart failure, but this in turn affects many other mechanisms. Failure to experience osmolarity changes will reduce the negative changes in terms of heart failure affected by osmolarity. A practical observable method is needed. CONCLUSIONS There is a possibility of using lens vacuoles in the follow-up of diabetic heart failure patients.
Collapse
Affiliation(s)
- Şahbender Koç
- University of Health Sciences, Keçiören Education and Training Hospital, Ankara, Turkey
| |
Collapse
|
19
|
Functional Expression of TRPV1 Ion Channel in the Canine Peripheral Blood Mononuclear Cells. Int J Mol Sci 2021; 22:ijms22063177. [PMID: 33804707 PMCID: PMC8003907 DOI: 10.3390/ijms22063177] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/12/2021] [Accepted: 03/17/2021] [Indexed: 12/25/2022] Open
Abstract
TRPV1, known as a capsaicin receptor, is the best-described transient receptor potential (TRP) ion channel. Recently, it was shown to be expressed by non-excitable cells such as lymphocytes. However, the data regarding the functional expression of the TRPV1 channel in the immune cells are often contradictory. In the present study, we performed a phylogenetical analysis of the canine TRP ion channels, we assessed the expression of TRPV1 in the canine peripheral blood mononuclear cells (PBMC) by qPCR and Western blot, and we determined the functionality of TRPV1 by whole-cell patch-clamp recordings and calcium assay. We found high expression of TRPV2, -M2, and -M7 in the canine PBMCs, while expression of TRPV1, -V4 and, -M5 was relatively low. We confirmed that TRPV1 is expressed on the protein level in the PBMC and it localizes in the plasma membrane. The whole-cell patch-clamp recording revealed that capsaicin application caused a significant increase in the current density. Similarly, the results from the calcium assay show a dose-dependent increase in intracellular calcium level in the presence of capsaicin that was partially abolished by capsazepine. Our study confirms the expression of TRPV1 ion channel on both mRNA and protein levels in the canine PBMC and indicates that the ion channel is functional.
Collapse
|
20
|
Nakazawa Y, Doki Y, Sugiyama Y, Kobayashi R, Nagai N, Morisita N, Endo S, Funakoshi-Tago M, Tamura H. Effect of Alpha-Glucosyl-Hesperidin Consumption on Lens Sclerosis and Presbyopia. Cells 2021; 10:cells10020382. [PMID: 33673261 PMCID: PMC7917927 DOI: 10.3390/cells10020382] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 11/16/2022] Open
Abstract
Presbyopia is characterized by a decline in the ability to accommodate the lens. The most commonly accepted theory for the onset of presbyopia is an age-related increase in the stiffness of the lens. However, the cause of lens sclerosis remains unclear. With age, water microcirculation in the lens could change because of an increase in intracellular pressure. In the lens, the intracellular pressure is controlled by the Transient Receptor Potential Vanilloid (TRPV) 1 and TRPV4 feedback pathways. In this study, we tried to elucidate that administration of α-glucosyl-hesperidin (G-Hsd), previously reported to prevent nuclear cataract formation, affects lens elasticity and the distribution of TRPV channels and Aquaporin (AQP) channels to meet the requirement of intracellular pressure. As a result, the mouse control lens was significantly toughened compared to both the 1% and 2% G-Hsd mouse lens treatments. The anti-oxidant levels in the lens and plasma decreased with age; however, this decrease could be nullified with either 1% or 2% G-Hsd treatment in a concentration- and exposure time-dependent manner. Moreover, G-Hsd treatment affected the TRPV4 distribution, but not TRPV1, AQP0, and AQP5, in the peripheral area and could maintain intracellular pressure. These findings suggest that G-Hsd has great potential as a compound to prevent presbyopia and/or cataract formation.
Collapse
Affiliation(s)
- Yosuke Nakazawa
- Faculty of Pharmacy, Keio University, 1-5-30 Shibako Minato-ku, Tokyo 105-8512, Japan; (Y.D.); (Y.S.); (R.K.); (M.F.-T.); (H.T.)
- Correspondence: ; Tel.: +81-3-5400-2689
| | - Yuri Doki
- Faculty of Pharmacy, Keio University, 1-5-30 Shibako Minato-ku, Tokyo 105-8512, Japan; (Y.D.); (Y.S.); (R.K.); (M.F.-T.); (H.T.)
| | - Yuki Sugiyama
- Faculty of Pharmacy, Keio University, 1-5-30 Shibako Minato-ku, Tokyo 105-8512, Japan; (Y.D.); (Y.S.); (R.K.); (M.F.-T.); (H.T.)
| | - Ryota Kobayashi
- Faculty of Pharmacy, Keio University, 1-5-30 Shibako Minato-ku, Tokyo 105-8512, Japan; (Y.D.); (Y.S.); (R.K.); (M.F.-T.); (H.T.)
| | - Noriaki Nagai
- Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka, Osaka 577-8502, Japan;
| | - Naoki Morisita
- R&D Division, Hayashibara Co., Ltd., Okayama 702-8006, Japan; (N.M.); (S.E.)
| | - Shin Endo
- R&D Division, Hayashibara Co., Ltd., Okayama 702-8006, Japan; (N.M.); (S.E.)
| | - Megumi Funakoshi-Tago
- Faculty of Pharmacy, Keio University, 1-5-30 Shibako Minato-ku, Tokyo 105-8512, Japan; (Y.D.); (Y.S.); (R.K.); (M.F.-T.); (H.T.)
| | - Hiroomi Tamura
- Faculty of Pharmacy, Keio University, 1-5-30 Shibako Minato-ku, Tokyo 105-8512, Japan; (Y.D.); (Y.S.); (R.K.); (M.F.-T.); (H.T.)
| |
Collapse
|
21
|
Yamamoto N, Takeda S, Hatsusaka N, Hiramatsu N, Nagai N, Deguchi S, Nakazawa Y, Takata T, Kodera S, Hirata A, Kubo E, Sasaki H. Effect of a Lens Protein in Low-Temperature Culture of Novel Immortalized Human Lens Epithelial Cells (iHLEC-NY2). Cells 2020; 9:cells9122670. [PMID: 33322631 PMCID: PMC7764252 DOI: 10.3390/cells9122670] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/27/2020] [Accepted: 12/11/2020] [Indexed: 11/24/2022] Open
Abstract
The prevalence of nuclear cataracts was observed to be significantly higher among residents of tropical and subtropical regions compared to those of temperate and subarctic regions. We hypothesized that elevated environmental temperatures may pose a risk of nuclear cataract development. The results of our in silico simulation revealed that in temperate and tropical regions, the human lens temperature ranges from 35.0 °C to 37.5 °C depending on the environmental temperature. The medium temperature changes during the replacement regularly in the cell culture experiment were carefully monitored using a sensor connected to a thermometer and showed a decrease of 1.9 °C, 3.0 °C, 1.7 °C, and 0.1 °C, after 5 min when setting the temperature of the heat plate device at 35.0 °C, 37.5 °C, 40.0 °C, and 42.5 °C, respectively. In the newly created immortalized human lens epithelial cell line clone NY2 (iHLEC-NY2), the amounts of RNA synthesis of αA crystallin, protein expression, and amyloid β (Aβ)1-40 secreted into the medium were increased at the culture temperature of 37.5 °C compared to 35.0 °C. In short-term culture experiments, the secretion of Aβ1-40 observed in cataracts was increased at 37.5 °C compared to 35.0 °C, suggesting that the long-term exposure to a high-temperature environment may increase the risk of cataracts.
Collapse
Affiliation(s)
- Naoki Yamamoto
- Department of Ophthalmology, Kanazawa Medical University, Ishikawa 920-0293, Japan; (N.Y.); (S.T.); (N.H.); (E.K.)
- Research Promotion and Support Headquarters, Fujita Health University, Aichi 470-1192, Japan;
| | - Shun Takeda
- Department of Ophthalmology, Kanazawa Medical University, Ishikawa 920-0293, Japan; (N.Y.); (S.T.); (N.H.); (E.K.)
| | - Natsuko Hatsusaka
- Department of Ophthalmology, Kanazawa Medical University, Ishikawa 920-0293, Japan; (N.Y.); (S.T.); (N.H.); (E.K.)
| | - Noriko Hiramatsu
- Research Promotion and Support Headquarters, Fujita Health University, Aichi 470-1192, Japan;
| | - Noriaki Nagai
- Faculty of Pharmacy, Kindai University, Osaka 577-8502, Japan; (N.N.); (S.D.)
| | - Saori Deguchi
- Faculty of Pharmacy, Kindai University, Osaka 577-8502, Japan; (N.N.); (S.D.)
| | - Yosuke Nakazawa
- Division of Hygienic Chemistry, Faculty of Pharmacy, Keio University, Tokyo 105-8512, Japan;
| | - Takumi Takata
- Radiation Biochemistry, Division of Radiation Life Science, Institute for Integrated Radiation and Nuclear Science, Kyoto University, Osaka 590-0494, Japan;
| | - Sachiko Kodera
- Department of Electrical and Mechanical Engineering, Nagoya Institute of Technology, Aichi 466-8555, Japan; (S.K.); (A.H.)
| | - Akimasa Hirata
- Department of Electrical and Mechanical Engineering, Nagoya Institute of Technology, Aichi 466-8555, Japan; (S.K.); (A.H.)
- Center of Biomedical Physics and Information Technology, Nagoya Institute of Technology, Aichi 466-8555, Japan
| | - Eri Kubo
- Department of Ophthalmology, Kanazawa Medical University, Ishikawa 920-0293, Japan; (N.Y.); (S.T.); (N.H.); (E.K.)
| | - Hiroshi Sasaki
- Department of Ophthalmology, Kanazawa Medical University, Ishikawa 920-0293, Japan; (N.Y.); (S.T.); (N.H.); (E.K.)
- Correspondence: ; Tel.: +81-762-286-2211
| |
Collapse
|
22
|
Guarino BD, Paruchuri S, Thodeti CK. The role of TRPV4 channels in ocular function and pathologies. Exp Eye Res 2020; 201:108257. [PMID: 32979394 DOI: 10.1016/j.exer.2020.108257] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/15/2022]
Abstract
Transient potential receptor vanilloid 4 (TRPV4) is an ion channel responsible for sensing osmotic and mechanical signals, which in turn regulates calcium signaling across cell membranes. TRPV4 is widely expressed throughout the body, and plays an important role in normal physiological function, as well as different pathologies, however, its role in the eye is not well known. In the eye, TRPV4 is expressed in various tissues, such as the retina, corneal epithelium, ciliary body, and the lens. In this review, we provide an overview on TRPV4 structure, activation, mutations, and summarize the current knowledge of TRPV4 function and signaling mechanisms in various locations throughout the eye, as well as its role in ocular diseases, such as glaucoma and diabetic retinopathy. Based on the available data, we highlight the therapeutic potential of TRPV4 as well as the shortcomings of current research. Finally, we provide future perspectives on the implications of targeting TRPV4 to treat various ocular pathologies.
Collapse
Affiliation(s)
- Brianna D Guarino
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44272, USA
| | | | - Charles K Thodeti
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44272, USA.
| |
Collapse
|
23
|
Petrova RS, Bavana N, Zhao R, Schey KL, Donaldson PJ. Changes to Zonular Tension Alters the Subcellular Distribution of AQP5 in Regions of Influx and Efflux of Water in the Rat Lens. Invest Ophthalmol Vis Sci 2020; 61:36. [PMID: 32945844 PMCID: PMC7509773 DOI: 10.1167/iovs.61.11.36] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/20/2020] [Indexed: 11/24/2022] Open
Abstract
Purpose The lens uses circulating fluxes of ions and water that enter the lens at both poles and exit at the equator to maintain its optical properties. We have mapped the subcellular distribution of the lens aquaporins (AQP0, AQP1, and AQP5) in these water influx and efflux zones and investigated how their membrane location is affected by changes in tension applied to the lens by the zonules. Methods Immunohistochemistry using AQP antibodies was performed on axial sections obtained from rat lenses that had been removed from the eye and then fixed or were fixed in situ to maintain zonular tension. Zonular tension was pharmacologically modulated by applying either tropicamide (increased) or pilocarpine (decreased). AQP labeling was visualized using confocal microscopy. Results Modulation of zonular tension had no effect on AQP1 or AQP0 labeling in either the water efflux or influx zones. In contrast, AQP5 labeling changed from membranous to cytoplasmic in response to both mechanical and pharmacologically induced reductions in zonular tension in both the efflux zone and anterior (but not posterior) influx zone associated with the lens sutures. Conclusions Altering zonular tension dynamically regulates the membrane trafficking of AQP5 in the efflux and anterior influx zones to potentially change the magnitude of circulating water fluxes in the lens.
Collapse
Affiliation(s)
- Rosica S. Petrova
- Department of Physiology, School of Medical Sciences, New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Nandini Bavana
- Department of Physiology, School of Medical Sciences, New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Rusin Zhao
- Department of Physiology, School of Medical Sciences, New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Kevin L. Schey
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, Tennessee, United States
| | - Paul J. Donaldson
- Department of Physiology, School of Medical Sciences, New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| |
Collapse
|
24
|
Nakazawa Y. Study of the Mechanisms of Maintaining the Transparency of the Lens and Treatment of Its Related Diseases for Making Anti-cataract and/or Anti-presbyopia Drugs. YAKUGAKU ZASSHI 2020; 140:1095-1099. [DOI: 10.1248/yakushi.20-00120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yosuke Nakazawa
- Division of Hygienic Chemistry, Faculty of Pharmacy, Keio University
| |
Collapse
|
25
|
Lapajne L, Lakk M, Yarishkin O, Gubeljak L, Hawlina M, Križaj D. Polymodal Sensory Transduction in Mouse Corneal Epithelial Cells. Invest Ophthalmol Vis Sci 2020; 61:2. [PMID: 32271891 PMCID: PMC7401707 DOI: 10.1167/iovs.61.4.2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Purpose Contact lenses, osmotic stressors, and chemical burns may trigger severe discomfort and vision loss by damaging the cornea, but the signaling mechanisms used by corneal epithelial cells (CECs) to sense extrinsic stressors are not well understood. We therefore investigated the mechanisms of swelling, temperature, strain, and chemical transduction in mouse CECs. Methods Intracellular calcium imaging in conjunction with electrophysiology, pharmacology, transcript analysis, immunohistochemistry, and bioluminescence assays of adenosine triphosphate (ATP) release were used to track mechanotransduction in dissociated CECs and epithelial sheets isolated from the mouse cornea. Results The transient receptor potential vanilloid (TRPV) transcriptome in the mouse corneal epithelium is dominated by Trpv4, followed by Trpv2, Trpv3, and low levels of Trpv1 mRNAs. TRPV4 protein was localized to basal and intermediate epithelial strata, keratocytes, and the endothelium in contrast to the cognate TRPV1, which was confined to intraepithelial afferents and a sparse subset of CECs. The TRPV4 agonist GSK1016790A induced cation influx and calcium elevations, which were abolished by the selective blocker HC067047. Hypotonic solutions, membrane strain, and moderate heat elevated [Ca2+]CEC with swelling- and temperature-, but not strain-evoked signals, sensitive to HC067047. GSK1016790A and swelling evoked calcium-dependent ATP release, which was suppressed by HC067027 and the hemichannel blocker probenecid. Conclusions These results demonstrate that cation influx via TRPV4 transduces osmotic and thermal but not strain inputs to CECs and promotes hemichannel-dependent ATP release. The TRPV4-hemichannel-ATP signaling axis might modulate corneal pain induced by excessive mechanical, osmotic, and chemical stimulation.
Collapse
|
26
|
Shahidullah M, Mandal A, Mathias RT, Gao J, Križaj D, Redmon S, Delamere NA. TRPV1 activation stimulates NKCC1 and increases hydrostatic pressure in the mouse lens. Am J Physiol Cell Physiol 2020; 318:C969-C980. [PMID: 32293931 PMCID: PMC7294325 DOI: 10.1152/ajpcell.00391.2019] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The porcine lens response to a hyperosmotic stimulus involves an increase in the activity of an ion cotransporter sodium-potassium/two-chloride cotransporter 1 (NKCC1). Recent studies with agonists and antagonists pointed to a mechanism that appears to depend on activation of transient receptor potential vanilloid 1 (TRPV1) ion channels. Here, we compare responses in lenses and cultured lens epithelium obtained from TRPV1-/- and wild type (WT) mice. Hydrostatic pressure (HP) in lens surface cells was determined using a manometer-coupled microelectrode approach. The TRPV1 agonist capsaicin (100 nM) caused a transient HP increase in WT lenses that peaked after ∼30 min and then returned toward baseline. Capsaicin did not cause a detectable change of HP in TRPV1-/- lenses. The NKCC inhibitor bumetanide prevented the HP response to capsaicin in WT lenses. Potassium transport was examined by measuring Rb+ uptake. Capsaicin increased Rb+ uptake in cultured WT lens epithelial cells but not in TRPV1-/- cells. Bumetanide, A889425, and the Akt inhibitor Akti prevented the Rb+ uptake response to capsaicin. The bumetanide-sensitive (NKCC-dependent) component of Rb+ uptake more than doubled in response to capsaicin. Capsaicin also elicited rapid (<2 min) NKCC1 phosphorylation in WT but not TRPV1-/- cells. HP recovery was shown to be absent in TRPV1-/- lenses exposed to hyperosmotic solution. Bumetanide and Akti prevented HP recovery in WT lenses exposed to hyperosmotic solution. Taken together, responses to capsaicin and hyperosmotic solution point to a functional role for TRPV1 channels in mouse lens. Lack of NKCC1 phosphorylation and Rb+ uptake responses in TRPV1-/- mouse epithelium reinforces the notion that a hyperosmotic challenge causes TRPV1-dependent NKCC1 activation. The results are consistent with a role for the TRPV1-activated signaling pathway leading to NKCC1 stimulation in lens osmotic homeostasis.
Collapse
Affiliation(s)
- Mohammad Shahidullah
- 1Department of Physiology, University of Arizona, Tucson, Arizona,2Department of Ophthalmology and Vision Science, University of Arizona, Tucson, Arizona
| | - Amritlal Mandal
- 1Department of Physiology, University of Arizona, Tucson, Arizona
| | - Richard T. Mathias
- 3Department of Physiology and Biophysics, Stony Brook University, Stony Brook, New York
| | - Junyuan Gao
- 3Department of Physiology and Biophysics, Stony Brook University, Stony Brook, New York
| | - David Križaj
- 4Department of Ophthalmology and Vision Science, University of Utah School of Medicine, Salt Lake City, Utah
| | - Sarah Redmon
- 4Department of Ophthalmology and Vision Science, University of Utah School of Medicine, Salt Lake City, Utah
| | - Nicholas A. Delamere
- 1Department of Physiology, University of Arizona, Tucson, Arizona,2Department of Ophthalmology and Vision Science, University of Arizona, Tucson, Arizona
| |
Collapse
|