1
|
Aszalós ZZ, Kolozsvári BL, Lénárt V, Pásztor D, Hassan Z, Surányi É, Chaker R, Fodor M. Sleep deprivation and corneal chronobiology: reevaluating overnight corneal changes. Sci Rep 2025; 15:801. [PMID: 39755902 DOI: 10.1038/s41598-024-84431-y] [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: 04/22/2024] [Accepted: 12/23/2024] [Indexed: 01/06/2025] Open
Abstract
This prospective cohort study is aimed to investigate circadian variations in corneal parameters, focusing on sleep-deprived subjects. Sixty-four healthy individuals (age range: 21-76 years) actively participated in this study, undergoing examinations at least five times within a 24-hour timeframe. The analysis encompassed keratometric parameters of the cornea's front (F) and back (B) surfaces, refractive power in flattest and steepest axes (K1, K2), astigmatism (Astig) and its axis (Axis), aspheric coefficient (Asph), corneal pachymetry values of thinnest corneal thickness (Pachy Min) and corneal thickness in the center of the pupil (Pachy Pupil), volume relative to the 3 and 10 mm corneal diagonal (Vol D3, Vol D10) and surface variance index (ISV). Circadian changes were assessed using a hierarchical, mixed-effects linear regression adjusted for age and night shift. A total of 1,636 measurements revealed significant circadian changes in various corneal parameters, including K1 F, K2 F/B, Astig F/B, Asph F/B, Pachy Min/Pupil, Vol D3/10, and ISV (p < 0.0001). Moreover, K1 B exhibited a significant circadian change (p = 0.0002), while Axis F/B remained unchanged. Notably, Corneal thickness peaked before 6 o'clock in the morning and reached its minimum after 12 o'clock. Contrary to previous notions linking corneal diurnal changes with eyelid closure during sleep, our study reveals that these changes persist in the absence of sleep. This research contributes valuable insights into the impact of sleep deprivation on corneal properties, warranting further investigations to deepen our understanding of daily variations in visual quality and guide the planning of refractive eye surgery interventions.
Collapse
Affiliation(s)
- Zsuzsa Zakarné Aszalós
- Department of Ophthalmology, Faculty of Medicine, University of Debrecen, Nagyerdei blvd. 98, Debrecen, 4012, Hungary
| | - Bence Lajos Kolozsvári
- Department of Ophthalmology, Faculty of Medicine, University of Debrecen, Nagyerdei blvd. 98, Debrecen, 4012, Hungary
| | - Vivien Lénárt
- Department of Ophthalmology, Faculty of Medicine, University of Debrecen, Nagyerdei blvd. 98, Debrecen, 4012, Hungary
| | - Dorottya Pásztor
- Department of Ophthalmology, Faculty of Medicine, University of Debrecen, Nagyerdei blvd. 98, Debrecen, 4012, Hungary
| | - Ziad Hassan
- Orbident Refractive Surgery and Medical Center, Nagyerdei krt. 98, Debrecen, 4012, Hungary
| | - Éva Surányi
- Department of Ophthalmology, Faculty of Medicine, University of Debrecen, Nagyerdei blvd. 98, Debrecen, 4012, Hungary
| | - Reda Chaker
- Department of Ophthalmology, Faculty of Medicine, University of Debrecen, Nagyerdei blvd. 98, Debrecen, 4012, Hungary
| | - Mariann Fodor
- Department of Ophthalmology, Faculty of Medicine, University of Debrecen, Nagyerdei blvd. 98, Debrecen, 4012, Hungary.
| |
Collapse
|
2
|
Wei R, Chen Q, Zheng Q, Reinach PS, Tan X, Pan C, Xu W, Tong L, Chen W. Epigenetic Activation of Circadian Clock Genes Elicits Inflammation in Experimental Murine Dry Eye. Ocul Immunol Inflamm 2024; 32:1180-1188. [PMID: 37163389 DOI: 10.1080/09273948.2023.2205525] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 03/28/2023] [Accepted: 04/17/2023] [Indexed: 05/12/2023]
Abstract
PURPOSE To explore whether circadian clock genes contribute to elicit inflammation in experimental dry eye (EDE). METHODS RNA sequencing analyzed mRNA expression patterns in EDE model. RT-qPCR and/or Western blot determined the expression of inflammatory factors and circadian genes during EDE. MethylTarget™ assays determined the promoter methylation levels of Per genes in vivo. Per2 or Per3 knockdown assessed their effects on inflammatory factors in vitro. RESULTS We utilized an intelligently controlled environmental system (ICES) to establish a mouse EDE model. The significant upregulated genes were enriched for circadian rhythms. Therein lied oscillatory and time-dependent upregulation of PER2 and PER3, as well as their promoter hypomethylation during EDE. Silencing PER2 or PER3 significantly decreased inflammatory factor expression and also reversed such increased inflammatory response in azacitidine (AZA) treatment in vitro model. CONCLUSIONS Our findings suggest that DNA methylation mediated the upregulation of PER2 and PER3, leading to inflammatory response in EDE.
Collapse
Affiliation(s)
- Ruifen Wei
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qianqian Chen
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qinxiang Zheng
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Peter S Reinach
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiying Tan
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chengjie Pan
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wei Xu
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Louis Tong
- Singapore Eye Research Institute, Singapore; Singapore National Eye Centre, Singapore; Duke-NUS Medical School, Singapore; Yong Loo Lin School of Medicine, Singapore; National University of Singapore, Singapore
| | - Wei Chen
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
3
|
Rolando M, Arnaldi D, Minervino A, Aragona P, Barabino S. Dry eye in mind: Exploring the relationship between sleep and ocular surface diseases. Eur J Ophthalmol 2024; 34:1128-1134. [PMID: 38111286 DOI: 10.1177/11206721231222063] [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] [Indexed: 12/20/2023]
Abstract
PURPOSE Dry Eye Disease (DED) is regarded as the most common ocular surface disease worldwide, entailing symptoms that have a major impact on the physical and psychological well-being of DED patients. In this context, the impact of sleep quality on DED has recently attracted attention. Indeed, although little is known about the mechanisms underlying the relationship between sleep and ocular surface diseases, recent evidence suggests that a reciprocal relationship exists between sleep quality and DED. Aim of the study was to investigate such relationship by means of both survey-based and instrumental analysis in a large population. PATIENTS AND METHODS The present cross-sectional study included 1182 DED patients who completed the Insomnia Severity Index (ISI) and the Ocular Surface Disease Index (OSDI) questionnaires. Moreover, tear break-up time (TBUT) and ocular surface staining (OSS) data of included patients were collected by physicians. RESULTS According to the findings of this study, in DED patients, the severity of dry eye symptoms and signs, assessed by OSDI score, TBUT, and ocular surface staining, is associated with more severe insomnia symptoms. Furthermore, higher severity of DED symptoms seems to be associated with the occurrence of nocturnal awakenings rather than with problems in falling asleep. CONCLUSIONS Present work contributes to the understanding of the complex relationship between DED and insomnia by showing that in a large population of DED patients, the more severe the insomnia, the more severe the DED symptoms and signs.
Collapse
Affiliation(s)
- Maurizio Rolando
- Ocular Surface and Dry Eye Center, University of Genoa & Is.Pre Oftalmica, Genoa, Italy
| | - Dario Arnaldi
- Department of Neuroscience, University of Genoa, Genoa, Italy
- Sleep Lab, Neurophysiopathology Unit, IRCSS Ospedale Policlinico San Martino, Genoa, Italy
| | | | - Pasquale Aragona
- Ophthalmology Clinic, Department of Biomedical Sciences, University of Messina, Messina, Italy
| | - Stefano Barabino
- Ocular Surface and Dry Eye Center, ASST Fatebenefratelli-Sacco, Ospedale Sacco-Università di Milano, Milan, Italy
| |
Collapse
|
4
|
Wang Y, Wang Q, Dou S, Zhou Q, Xie L. Sleep deprivation induces corneal endothelial dysfunction by downregulating Bmal1. BMC Ophthalmol 2024; 24:268. [PMID: 38907352 PMCID: PMC11191275 DOI: 10.1186/s12886-024-03524-4] [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: 04/03/2023] [Accepted: 06/12/2024] [Indexed: 06/23/2024] Open
Abstract
BACKGROUND Sleep deprivation (SD) is a common public health problem that contributes to various physiological disorders and increases the risk of ocular diseases. However, whether sleep loss can damage corneal endothelial function remains unclear. This study aimed to determine the effect and possible mechanism of SD on the corneal endothelium. METHODS Male C57BL/6J mice were subjected to establish SD models. After 10 days, quantitative RT-PCR (qRT-PCR) and western blot or immunostaining for the expression levels of zonula occludens-1 (ZO-1), ATPase Na+/K + transporting subunit alpha 1 (Atp1a1), and core clock genes in the corneal endothelium were evaluated. Reactive oxygen species staining and mitochondrial abundance characterized the mitochondrial function. The regulatory role of Bmal1 was confirmed by specifically knocking down or overexpressing basic helix-loop-helix ARNT like 1 protein (Bmal1) in vivo. In vitro, a mitochondrial stress test was conducted on cultured human corneal endothelial cells upon Bmal1 knockdown. RESULTS SD damaged the barrier and pump functions of mouse corneal endothelium, accompanied by mitochondrial dysfunction. Interestingly, SD dramatically downregulated the core clock gene Bmal1 expression level. Bmal1 knockdown disrupted corneal endothelial function, while overexpression of Bmal1 ameliorated the dysfunction induced by SD. Mitochondrial bioenergetic deficiency mediated by Bmal1 was an underlying mechanism for SD induced corneal endothelial dysfunction. CONCLUSION The downregulation of Bmal1 expression caused by SD led to corneal endothelial dysfunction via impairing mitochondrial bioenergetics. Our findings offered insight into how SD impairs the physiological function of the corneal endothelium and expanded the understanding of sleep loss leading to ocular diseases.
Collapse
Affiliation(s)
- Yani Wang
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, 5 Yan er dao Road, Qingdao, 266071, China
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong First Medical University, Shandong, China
- School of ophthalmology, Shandong First Medical University, Shandong, China
| | - Qun Wang
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, 5 Yan er dao Road, Qingdao, 266071, China
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong First Medical University, Shandong, China
- School of ophthalmology, Shandong First Medical University, Shandong, China
| | - Shengqian Dou
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, 5 Yan er dao Road, Qingdao, 266071, China
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong First Medical University, Shandong, China
- School of ophthalmology, Shandong First Medical University, Shandong, China
| | - Qingjun Zhou
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, 5 Yan er dao Road, Qingdao, 266071, China
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong First Medical University, Shandong, China
- School of ophthalmology, Shandong First Medical University, Shandong, China
| | - Lixin Xie
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, 5 Yan er dao Road, Qingdao, 266071, China.
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong First Medical University, Shandong, China.
- School of ophthalmology, Shandong First Medical University, Shandong, China.
| |
Collapse
|
5
|
Xue Y, Xu P, Hu Y, Liu S, Yan R, Liu S, Li Y, Liu J, Fu T, Li Z. Stress systems exacerbate the inflammatory response after corneal abrasion in sleep-deprived mice via the IL-17 signaling pathway. Mucosal Immunol 2024; 17:323-345. [PMID: 38428739 DOI: 10.1016/j.mucimm.2024.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/22/2024] [Accepted: 02/23/2024] [Indexed: 03/03/2024]
Abstract
Sleep deprivation (SD) has a wide range of adverse health effects. However, the mechanisms by which SD influences corneal pathophysiology and its post-wound healing remain unclear. This study aimed to examine the basic physiological characteristics of the cornea in mice subjected to SD and determine the pathophysiological response to injury after corneal abrasion. Using a multi-platform water environment method as an SD model, we found that SD leads to disturbances of corneal proliferative, sensory, and immune homeostasis as well as excessive inflammatory response and delayed repair after corneal abrasion by inducing hyperactivation of the sympathetic nervous system and hypothalamic-pituitary-adrenal axis. Pathophysiological changes in the cornea mainly occurred through the activation of the IL-17 signaling pathway. Blocking both adrenergic and glucocorticoid synthesis and locally neutralizing IL-17A significantly improved corneal homeostasis and the excessive inflammatory response and delay in wound repair following corneal injury in SD-treated mice. These results indicate that optimal sleep quality is essential for the physiological homeostasis of the cornea and its well-established repair process after injury. Additionally, these observations provide potential therapeutic targets to ameliorate SD-induced delays in corneal wound repair by inhibiting or blocking the activation of the stress system and its associated IL-17 signaling pathway.
Collapse
Affiliation(s)
- Yunxia Xue
- International Ocular Surface Research Center, Institute of Ophthalmology and Key Laboratory for Regenerative Medicine, Jinan University Medical School, Guangzhou, China; Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Pengyang Xu
- International Ocular Surface Research Center, Institute of Ophthalmology and Key Laboratory for Regenerative Medicine, Jinan University Medical School, Guangzhou, China; Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China; Department of Pathology, Nanyang Second General Hospital, Nanyang City, Henan, China
| | - Yu Hu
- International Ocular Surface Research Center, Institute of Ophthalmology and Key Laboratory for Regenerative Medicine, Jinan University Medical School, Guangzhou, China
| | - Sijing Liu
- International Ocular Surface Research Center, Institute of Ophthalmology and Key Laboratory for Regenerative Medicine, Jinan University Medical School, Guangzhou, China; Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Ruyu Yan
- International Ocular Surface Research Center, Institute of Ophthalmology and Key Laboratory for Regenerative Medicine, Jinan University Medical School, Guangzhou, China; Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Shutong Liu
- International Ocular Surface Research Center, Institute of Ophthalmology and Key Laboratory for Regenerative Medicine, Jinan University Medical School, Guangzhou, China
| | - Yan Li
- International Ocular Surface Research Center, Institute of Ophthalmology and Key Laboratory for Regenerative Medicine, Jinan University Medical School, Guangzhou, China; Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jun Liu
- International Ocular Surface Research Center, Institute of Ophthalmology and Key Laboratory for Regenerative Medicine, Jinan University Medical School, Guangzhou, China; Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Ting Fu
- International Ocular Surface Research Center, Institute of Ophthalmology and Key Laboratory for Regenerative Medicine, Jinan University Medical School, Guangzhou, China; Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Zhijie Li
- International Ocular Surface Research Center, Institute of Ophthalmology and Key Laboratory for Regenerative Medicine, Jinan University Medical School, Guangzhou, China; Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China.
| |
Collapse
|
6
|
Woodard G, Rosado JA, Li H. The physiological role of TRP channels in sleep and circadian rhythm. J Cell Mol Med 2024; 28:e18274. [PMID: 38676362 PMCID: PMC11053353 DOI: 10.1111/jcmm.18274] [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: 04/30/2023] [Revised: 10/24/2023] [Accepted: 01/05/2024] [Indexed: 04/28/2024] Open
Abstract
TRP channels, are non-specific cationic channels that are involved in multiple physiological processes that include salivation, cellular secretions, memory extinction and consolidation, temperature, pain, store-operated calcium entry, thermosensation and functionality of the nervous system. Here we choose to look at the evidence that decisively shows how TRP channels modulate human neuron plasticity as it relates to the molecular neurobiology of sleep/circadian rhythm. There are numerous model organisms of sleep and circadian rhythm that are the results of the absence or genetic manipulation of the non-specific cationic TRP channels. Drosophila and mice that have had their TRP channels genetically ablated or manipulated show strong evidence of changes in sleep duration, sleep activity, circadian rhythm and response to temperature, noxious odours and pattern of activity during both sleep and wakefulness along with cardiovascular and respiratory function during sleep. Indeed the role of TRP channels in regulating sleep and circadian rhythm is very interesting considering the parallel roles of TRP channels in thermoregulation and thermal response with concomitant responses in growth and degradation of neurites, peripheral nerves and neuronal brain networks. TRP channels provide evidence of an ability to create, regulate and modify our sleep and circadian rhythm in a wide array of physiological and pathophysiological conditions. In the current review, we summarize previous results and novel recent advances in the understanding of calcium ion entry via TRP channels in different sleep and circadian rhythm conditions. We discuss the role of TRP channels in sleep and circadian disorders.
Collapse
Affiliation(s)
- Geoffrey Woodard
- Department of PsychiatryUniformed Services University of Health SciencesBethesdaMarylandUSA
| | - Juan A. Rosado
- Department of PhysiologyUniversity of ExtremaduraCaceresSpain
| | - He Li
- Department of PsychiatryUniformed Services University of Health SciencesBethesdaMarylandUSA
| |
Collapse
|
7
|
Tang L, Zhang H, Liao Y, Zhou S, Yang Y, Zhang M, Guo Y, Xie T, Chen S, Ouyang W, Lin X, Wang S, Huang C, Zhang M, Zhuang J, Zhao J, Zhang R, Zhang C, Jin Z, Hu J, Liu Z. Chronic Sleep Deprivation Impairs Visual Functions via Oxidative Damage in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:307-320. [PMID: 38245252 DOI: 10.1016/j.ajpath.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 10/16/2023] [Accepted: 11/06/2023] [Indexed: 01/22/2024]
Abstract
Sleep deprivation (SD) is a global public health burden, and has a detrimental role in the nervous system. Retina is an important part of the central nervous system; however, whether SD affects retinal structures and functions remains largely unknown. Herein, chronic SD mouse model indicated that loss of sleep for 4 months could result in reductions in the visual functions, but without obvious morphologic changes of the retina. Ultrastructural analysis by transmission electron microscope revealed the deterioration of mitochondria, which was accompanied with the decrease of multiple mitochondrial proteins in the retina. Mechanistically, oxidative stress was provoked by chronic SD, which could be ameliorated after rest, and thus restore retinal homeostasis. Moreover, the supplementation of two antioxidants, α-lipoic acid and N-acetyl-l-cysteine, could reduce retinal reactive oxygen species, repair damaged mitochondria, and, as a result, improve the retinal functions. Overall, this work demonstrated the essential roles of sleep in maintaining the integrity and health of the retina. More importantly, it points towards supplementation of antioxidants as an effective intervention strategy for people experiencing sleep shortages.
Collapse
Affiliation(s)
- Liying Tang
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Department of Ophthalmology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Houjian Zhang
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Department of Ophthalmology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Yi Liao
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Shengmei Zhou
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yaqiong Yang
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Mouxin Zhang
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yuli Guo
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Department of Ophthalmology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Tingyu Xie
- Department of Ophthalmology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Shikun Chen
- Department of Ophthalmology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Weijie Ouyang
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xiang Lin
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Shaopan Wang
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Caihong Huang
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Minjie Zhang
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jingbin Zhuang
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jiankai Zhao
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Rongrong Zhang
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Changjun Zhang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Laboratory, Beijing, China
| | - Zibing Jin
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Laboratory, Beijing, China
| | - Jiaoyue Hu
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Department of Ophthalmology, The First Affiliated Hospital of University of South China, Hengyang, China; Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.
| | - Zuguo Liu
- Xiamen University-affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Department of Ophthalmology, The First Affiliated Hospital of University of South China, Hengyang, China; Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.
| |
Collapse
|
8
|
Preoteasa LD, Preoteasa D. Assessment of the prevalence and risk factors for dry eye symptoms among Romanian medical students using the ocular surface disease index - a cross-sectional study. BMC Ophthalmol 2024; 24:12. [PMID: 38182974 PMCID: PMC10768241 DOI: 10.1186/s12886-023-03260-1] [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: 04/15/2023] [Accepted: 12/07/2023] [Indexed: 01/07/2024] Open
Abstract
BACKGROUND This study aims to assess how Romanian medical students suffer from dry eye disease symptoms, establish the prevalence and severity of dry eye (DE) symptoms and identify potential risk factors. METHODS An analytical, cross-sectional study was conducted on students from "Carol Davila" University of Medicine, Romania, after the final examination period of July 2022. The OSDI score (Ocular Surface Disease Index©) was applied in an online survey. The study adopted the standards used by other authors, who defined symptomatic DED as an OSDI score greater than 12. The chi-square test was used to establish statistical significance at a cutoff value of p < 0.05. The predictive model was created using linear logistic regression analysis. The goodness of fit of the logistic regression model was assessed using the Hosmer-Lemeshow test. When the severity outcome had a nominal categorical form, multinomial regression analysis with normal subjects as a reference was performed. The distribution of the severe type of symptomatology across sex categories and years of study was analyzed using a nonparametric test (Independent-Samples Kruskal-Wallis Test). RESULTS A total of 274 answers were received from 81.4% females and 18.6% males with a response rate of 35.58%. The mean age was 22.7 years ± 1.66 with an age range between 20 and 25 years old. Using the OSDI score, we established that the overall prevalence of DE symptoms was 83.6% (95%CI: 79.6%, 88%), with an 85.2% (95%CI: 80.5%, 89.8%) prevalence in females and 76.5% (95%CI: 65%,88%) in males. The severe form of DE was the most prevalent, regardless of the study year or sex. Increased screen time (p-value < 0.05) and non-smokers (p-value < 0.05) were proven risk factors. The predictive model which includes the explanatory variables (sex, contact lens wearers, smoking, oral contraceptives, screen time) proved an 84.7% predictability for symptomatic DE and was able to better predict the dependent variable than the intercept model only (p-value < 0.05). Smoking (p = 0.002) and screen time (p = 0.009) preserved their significance in the multinominal regression as well. CONCLUSIONS This is the first study to report the epidemiology of DE symptoms among Romanian medical students. OSDI revealed a high prevalence of symptomatic DE in medical students. Screen time, although not the only factor, likely plays a role in exacerbating the disease. This information can be used to inform healthcare policies, establish occupational health guidelines, and implement preventive measures for individuals in similar high-stress academic or professional environments.
Collapse
Affiliation(s)
- Laura Denisa Preoteasa
- Carol Davila University of Medicine and Pharmacy, Dionisie Lupu street, no 37, Bucharest, 030167, Romania.
- Department of Ophthalmology, Clinical Emergency Eye Hospital, Bucharest, Romania.
- Onioptic Hospital, Craiova, Romania.
| | | |
Collapse
|
9
|
Chien LN, Chou YI, Tsai YJ, Sung LC, Liu HY, Lin MY, Wang TJ, Lin IC. Association of lipid-lowering agent use and dry eye disease: A nationwide matched case-control study in Taiwan, 2002-2016. Acta Ophthalmol 2023; 101:729-736. [PMID: 36942369 DOI: 10.1111/aos.15656] [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: 09/20/2022] [Revised: 02/02/2023] [Accepted: 02/26/2023] [Indexed: 03/23/2023]
Abstract
PURPOSE The purpose of this study is to evaluate the association between lipid-lowering agent use and the risks of diagnosed dry eye disease (DED). METHODS This retrospective, case-control study included 780 786 patients who received lipid-lowering agents in 2002-2016, of which 17 409 were newly diagnosed with DED during a ≥2-year follow-up period. These patients were matched 1:4 with control participants for age, sex, and comorbidities. Separate odds ratios (OR) were calculated for DED and each of statin and fibrate use. RESULTS Statin users had significantly higher odds of DED (adjusted OR = 1.12; 95% confidence interval (CI) = 1.08-1.16, p < 0.0001) than nonusers. Fibrate users did not show higher odds of DED than nonusers (adjusted OR = 1.04; 95% CI = 0.99-1.10, p = 0.125). The lipophilic statin users did not show higher odds of DED compared with the hydrophilic statin users (adjusted OR = 0.99, 95% CI = 0.93-1.06, p = 0.729). Among statin users, the odds of DED did not differ significantly between patients receiving statin therapy for >180 days vs. ≤90 days or patients receiving statin therapy for 91-180 days vs. ≤90 days (adjusted OR = 1.00, p = 0.922; adjusted OR = 0.94, p = 0.541, respectively). The odds of DED were not statistically different among patients receiving low-intensity, moderate-intensity, and high-intensity of statin therapy. CONCLUSIONS Patients receiving statin therapy had a higher DED risk than patients not receiving statin therapy. The type of statin, the duration, and the intensity of statin use were not significantly associated with DED risks. Further studies are required to identify the relevant factors related to DED risks with statin.
Collapse
Affiliation(s)
- Li-Nien Chien
- Institute of Health and Welfare Policy, National Yang Ming Chios Tung University, Taipei, Taiwan
- Graduate Institute of Data Science, College of Management, Taipei Medical University, Taipei, Taiwan
| | - Yun-I Chou
- Department of Dermatology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yuan-Jen Tsai
- Department of Family Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Li-Chin Sung
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Primary Care Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
| | - Hung-Yi Liu
- Data Analytics and Statistics Center, Taipei Medical University, Taipei, Taiwan
- School of Health Care Administration, College of Management, Taipei Medical University, Taipei, Taiwan
| | - Meng-Yin Lin
- Department of Ophthalmology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Ophthalmology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tsung-Jen Wang
- Department of Ophthalmology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Ophthalmology, Taipei Medical University Hospital, Taipei, Taiwan
| | - I-Chan Lin
- Department of Ophthalmology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Ophthalmology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Ophthalmology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
10
|
Galor A, Britten-Jones AC, Feng Y, Ferrari G, Goldblum D, Gupta PK, Merayo-Lloves J, Na KS, Naroo SA, Nichols KK, Rocha EM, Tong L, Wang MTM, Craig JP. TFOS Lifestyle: Impact of lifestyle challenges on the ocular surface. Ocul Surf 2023; 28:262-303. [PMID: 37054911 DOI: 10.1016/j.jtos.2023.04.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 04/07/2023] [Indexed: 04/15/2023]
Abstract
Many factors in the domains of mental, physical, and social health have been associated with various ocular surface diseases, with most of the focus centered on aspects of dry eye disease (DED). Regarding mental health factors, several cross-sectional studies have noted associations between depression and anxiety, and medications used to treat these disorders, and DED symptoms. Sleep disorders (both involving quality and quantity of sleep) have also been associated with DED symptoms. Under the domain of physical health, several factors have been linked to meibomian gland abnormalities, including obesity and face mask wear. Cross-sectional studies have also linked chronic pain conditions, specifically migraine, chronic pain syndrome and fibromyalgia, to DED, principally focusing on DED symptoms. A systematic review and meta-analysis reviewed available data and concluded that various chronic pain conditions increased the risk of DED (variably defined), with odds ratios ranging from 1.60 to 2.16. However, heterogeneity was noted, highlighting the need for additional studies examining the impact of chronic pain on DED signs and subtype (evaporative versus aqueous deficient). With respect to societal factors, tobacco use has been most closely linked to tear instability, cocaine to decreased corneal sensitivity, and alcohol to tear film disturbances and DED symptoms.
Collapse
Affiliation(s)
- Anat Galor
- Bascom Palmer Eye Institute, University of Miami, Miami, FL, USA; Surgical Services, Miami Veterans Administration, Miami, FL, USA.
| | - Alexis Ceecee Britten-Jones
- Department of Optometry and Vision Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Victoria, Australia
| | - Yun Feng
- Department of Ophthalmology, Peking University Eye Center, Peking University Third Hospital, Beijing, China
| | - Giulio Ferrari
- Cornea and Ocular Surface Unit, Eye Repair Lab, San Raffaele Scientific Institute, Milan, Italy
| | - David Goldblum
- Pallas-Kliniken, Olten, Bern, Zurich, Switzerland; University of Basel, Basel, Switzerland
| | - Preeya K Gupta
- Triangle Eye Consultants, Raleigh, NC, USA; Department of Ophthalmology, Tulane University, New Orleans, LA, USA
| | - Jesus Merayo-Lloves
- Instituto Universitario Fernandez-Vega, Universidad de Oviedo, Principality of Asturias, Spain
| | - Kyung-Sun Na
- Department of Ophthalmology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Shehzad A Naroo
- College of Health and Life Sciences, Aston University, Birmingham, UK
| | - Kelly K Nichols
- School of Optometry, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Eduardo M Rocha
- Department of Ophthalmology, Othorynolaringology and Head & Neck Surgery, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Louis Tong
- Cornea and External Eye Disease Service, Singapore National Eye Center, Ocular Surface Research Group, Singapore Eye Research Institute, Eye Academic Clinical Program, Duke-National University of Singapore, Singapore
| | - Michael T M Wang
- Department of Ophthalmology, New Zealand National Eye Centre, The University of Auckland, Auckland, New Zealand
| | - Jennifer P Craig
- Department of Ophthalmology, New Zealand National Eye Centre, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
11
|
Zhou T, Yan K, Zhang Y, Zhu L, Liao Y, Zheng X, Chen Y, Li X, Liu Z, Zhang Z. Fenofibrate suppresses corneal neovascularization by regulating lipid metabolism through PPARα signaling pathway. Front Pharmacol 2022; 13:1000254. [PMID: 36588740 PMCID: PMC9800935 DOI: 10.3389/fphar.2022.1000254] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
Purpose: The purpose of this study was to explore the potential underlying mechanism of anti-vascular effects of peroxisome proliferator-activated receptor α (PPARα) agonist fenofibrate against corneal neovascularization (CNV) through the changes of lipid metabolism during CNV. Methods: A suture-induced CNV model was established and the clinical indications were evaluated from day 1 to day 7. Treatments of vehicle and fenofibrate were performed for 5 days after suture and the CNV areas were compared among the groups. The eyeballs were collected for histological analysis, malondialdehyde (MDA) measurement, terminal deoxynucleotidyl transferase 2'-deoxyuridine 5'-triphosphate nick end labeling (TUNEL) staining, western blot, quantitative real-time PCR (qRT-PCR) assays and immunohistochemical (IHC) staining to elucidate pathological changes and the underlying mechanism. Results: Lipi-Green staining and MDA measurement showed that lipid deposition and peroxidation were increased in the CNV cornea while the expression of long-chain acyl-coenzyme A synthetase 1 (ACSL1), carnitine palmitoyltransterase 1A(CPT1A) and medium-chain acyl-coenzyme A dehydrogenase (ACADM), which are key enzymes of fatty acid β-oxidation (FAO) and targeted genes of peroxisome proliferator-activated receptor alpha (PPARα) pathway, were decreased in CNV cornea. Fenofibrate suppressed lipid accumulation and peroxidation damage in the CNV cornea. Fenofibrate upregulated the expression levels of PPARα, ACSL1, CPT1A, and ACADM compared with vehicle group. IHC staining indicated that fenofibrate also decreased the expression of VEGFa, VEGFc, TNFα, IL1β and CD68. Conclusion: Disorder of lipid metabolism may be involved in the formation of suture-induced CNV and fenofibrate played anti-neovascularization and anti-inflammatory roles on cornea by regulating the key enzymes of lipid metabolism and ameliorating lipid peroxidation damage of cornea through PPARα signaling pathway.
Collapse
Affiliation(s)
- Tong Zhou
- Eye Institute and Affiliated Xiamen Eye Center of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China,Department of Pharmacy, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China,Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Fujian Provincial Key Laboratory of Corneal and Ocular Surface Diseases, Xiamen, China
| | - Ke Yan
- The First Affiliated Hospital, Department of Ophthalmology, Hengyang Medical School, University of South China, Hengyang, China
| | - Yuhan Zhang
- Eye Institute and Affiliated Xiamen Eye Center of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China,Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Fujian Provincial Key Laboratory of Corneal and Ocular Surface Diseases, Xiamen, China
| | - Linfangzi Zhu
- Eye Institute and Affiliated Xiamen Eye Center of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China,Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Fujian Provincial Key Laboratory of Corneal and Ocular Surface Diseases, Xiamen, China
| | - Yi Liao
- Eye Institute and Affiliated Xiamen Eye Center of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China,Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Fujian Provincial Key Laboratory of Corneal and Ocular Surface Diseases, Xiamen, China
| | - Xiaoxiang Zheng
- Eye Institute and Affiliated Xiamen Eye Center of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China,Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Fujian Provincial Key Laboratory of Corneal and Ocular Surface Diseases, Xiamen, China
| | - Yongxiong Chen
- Eye Institute and Affiliated Xiamen Eye Center of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China,Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Fujian Provincial Key Laboratory of Corneal and Ocular Surface Diseases, Xiamen, China
| | - Xiaoxin Li
- Eye Institute and Affiliated Xiamen Eye Center of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China,Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Fujian Provincial Key Laboratory of Corneal and Ocular Surface Diseases, Xiamen, China,Department of Ophthalmology and Clinical Centre of Optometry, Peking University People’s Hospital, Beijing, China,*Correspondence: Zhaoqiang Zhang, ; Zuguo Liu, ; Xiaoxin Li,
| | - Zuguo Liu
- Eye Institute and Affiliated Xiamen Eye Center of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China,Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Fujian Provincial Key Laboratory of Corneal and Ocular Surface Diseases, Xiamen, China,The First Affiliated Hospital, Department of Ophthalmology, Hengyang Medical School, University of South China, Hengyang, China,*Correspondence: Zhaoqiang Zhang, ; Zuguo Liu, ; Xiaoxin Li,
| | - Zhaoqiang Zhang
- Eye Institute and Affiliated Xiamen Eye Center of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China,Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Fujian Provincial Key Laboratory of Corneal and Ocular Surface Diseases, Xiamen, China,*Correspondence: Zhaoqiang Zhang, ; Zuguo Liu, ; Xiaoxin Li,
| |
Collapse
|
12
|
Transient Receptor Potential Channels: Important Players in Ocular Pain and Dry Eye Disease. Pharmaceutics 2022; 14:pharmaceutics14091859. [PMID: 36145607 PMCID: PMC9506338 DOI: 10.3390/pharmaceutics14091859] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/26/2022] [Accepted: 08/26/2022] [Indexed: 11/20/2022] Open
Abstract
Dry eye disease (DED) is a multifactorial disorder in which the eyes respond to minor stimuli with abnormal sensations, such as dryness, blurring, foreign body sensation, discomfort, irritation, and pain. Corneal pain, as one of DED’s main symptoms, has gained recognition due to its increasing prevalence, morbidity, and the resulting social burden. The cornea is the most innervated tissue in the body, and the maintenance of corneal integrity relies on a rich density of nociceptors, such as polymodal nociceptor neurons, cold thermoreceptor neurons, and mechano-nociceptor neurons. Their sensory responses to different stimulating forces are linked to the specific expression of transient receptor potential (TRP) channels. TRP channels are a group of unique ion channels that play important roles as cellular sensors for various stimuli. These channels are nonselective cation channels with variable Ca2+ selectivity. TRP homologs are a superfamily of 28 different members that are subdivided into 7 different subfamilies based on differences in sequence homology. Many of these subtypes are expressed in the eye on both neuronal and non-neuronal cells, where they affect various stress-induced regulatory responses essential for normal vision maintenance. This article reviews the current knowledge about the expression, function, and regulation of TRPs in ocular surface tissues. We also describe their implication in DED and ocular pain. These findings contribute to evidence suggesting that drug-targeting TRP channels may be of therapeutic benefit in the clinical setting of ocular pain.
Collapse
|
13
|
Sahu M, Tripathi R, Jha NK, Jha SK, Ambasta RK, Kumar P. Cross talk mechanism of disturbed sleep patterns in neurological and psychological disorders. Neurosci Biobehav Rev 2022; 140:104767. [PMID: 35811007 DOI: 10.1016/j.neubiorev.2022.104767] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/20/2022] [Accepted: 07/01/2022] [Indexed: 11/25/2022]
Abstract
The incidence and prevalence of sleep disorders continue to increase in the elderly populace, particularly those suffering from neurodegenerative and neuropsychiatric disorders. This not only affects the quality of life but also accelerates the progression of the disease. There are many reasons behind sleep disturbances in such patients, for instance, medication use, nocturia, obesity, environmental factors, nocturnal motor disturbances and depressive symptoms. This review focuses on the mechanism and effects of sleep dysfunction in neurodegenerative and neuropsychiatric disorders. Wherein we discuss disturbed circadian rhythm, signaling cascade and regulation of genes during sleep deprivation. Moreover, we explain the perturbation in brainwaves during disturbed sleep and the ocular perspective of neurodegenerative and neuropsychiatric manifestations in sleep disorders. Further, as the pharmacological approach is often futile and carries side effects, therefore, the non-pharmacological approach opens newer possibilities to treat these disorders and widens the landscape of treatment options for patients.
Collapse
Affiliation(s)
- Mehar Sahu
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India
| | - Rahul Tripathi
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET) Sharda University, UP, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET) Sharda University, UP, India.
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India.
| |
Collapse
|
14
|
Giannos P, Prokopidis K, Forbes SC, Celoch K, Candow DG, Tartar JL. Gene Expression Changes of Murine Cortex Homeostasis in Response to Sleep Deprivation Hint Dysregulated Aging-like Transcriptional Responses. Brain Sci 2022; 12:825. [PMID: 35884632 PMCID: PMC9313387 DOI: 10.3390/brainsci12070825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/16/2022] [Accepted: 06/17/2022] [Indexed: 12/04/2022] Open
Abstract
Sleep deprivation leads to the deterioration in the physiological functioning of the brain, cognitive decline, and many neurodegenerative diseases, all of which progress with advancing age. Sleep insufficiency and impairments in cognitive function are characterized by progressive neuronal losses in the cerebral cortex. In this study, we analyze gene expression profiles following sleep-deprived murine models and circadian matched controls to identify genes that might underlie cortical homeostasis in response to sleep deprivation. Screening of the literature resulted in three murine (Mus musculus) gene expression datasets (GSE6514, GSE78215, and GSE33491) that included cortical tissue biopsies from mice that are sleep deprived for 6 h (n = 15) and from circadian controls that are left undisturbed (n = 15). Cortical differentially expressed genes are used to construct a network of encoded proteins that are ranked based on their interactome according to 11 topological algorithms. The analysis revealed three genes-NFKBIA, EZR, and SGK1-which exhibited the highest multi-algorithmic topological significance. These genes are strong markers of increased brain inflammation, cytoskeletal aberrations, and glucocorticoid resistance, changes that imply aging-like transcriptional responses during sleep deprivation in the murine cortex. Their potential role as candidate markers of local homeostatic response to sleep loss in the murine cortex warrants further experimental validation.
Collapse
Affiliation(s)
- Panagiotis Giannos
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, South Kensington, London SW7 2AZ, UK
- Society of Meta-Research and Biomedical Innovation, London W12 0BZ, UK;
| | - Konstantinos Prokopidis
- Society of Meta-Research and Biomedical Innovation, London W12 0BZ, UK;
- Department of Musculoskeletal Biology, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L69 3BX, UK
| | - Scott C. Forbes
- Department of Physical Education Studies, Faculty of Education, Brandon University, Brandon, MB R7A 6A9, Canada;
| | - Kamil Celoch
- Department of Psychology and Neuroscience, Nova Southeastern University, Fort Lauderdale, FL 33314, USA; (K.C.); (J.L.T.)
| | - Darren G. Candow
- Faculty of Kinesiology and Health Studies, University of Regina, Regina, SK S4S 0A2, Canada;
| | - Jaime L. Tartar
- Department of Psychology and Neuroscience, Nova Southeastern University, Fort Lauderdale, FL 33314, USA; (K.C.); (J.L.T.)
| |
Collapse
|
15
|
Guo X, Dang W, Li N, Wang Y, Sun D, Nian H, Wei R. PPAR-α Agonist Fenofibrate Ameliorates Sjögren Syndrome-Like Dacryoadenitis by Modulating Th1/Th17 and Treg Cell Responses in NOD Mice. Invest Ophthalmol Vis Sci 2022; 63:12. [PMID: 35687344 PMCID: PMC9202336 DOI: 10.1167/iovs.63.6.12] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To investigate the effects and mechanisms of fenofibrate, a synthetic ligand of peroxisome proliferator-activated receptor α (PPAR-α), on autoimmune dacryoadenitis in a mouse model of Sjögren syndrome (SS) dry eye. Methods Male nonobese diabetic (NOD) mice were fed chow with or without 0.03% fenofibrate for 8 weeks, and clinical scores were determined by assessing tear secretion, fluorescein, and hematoxylin and eosin staining. Intracellular IFN-γ, IL-17, and Foxp3 in CD4+ T cells were measured by flow cytometry. The expressions of Th1, Th17, and Treg cell-related transcription factors and cytokines were detected by real-time PCR. The levels of PPAR-α and liver X receptor β (LXR-β) were detected with real-time PCR and Western blotting. Results Fenofibrate efficiently diminished the lymphocytic inflammation in lacrimal glands (LGs), increased tear secretion, and decreased corneal fluorescein staining in NOD mice. Meanwhile, treatment of fenofibrate evidently reduced the proportion of Th1 and Th17 cells and increased the proportion of Treg cells in vivo and vitro, together with decreased expression of T-bet, IFN-γ, RORγt, and IL-17, as well as increased expression of Foxp3 and TGF-β1 in LGs. Furthermore, fenofibrate significantly upregulated the expressions of PPAR-α and LXR-β at the protein and mRNA levels. Conclusions Fenofibrate potently attenuated LG inflammation in a model of autoimmune dry eye, and this effect might partially result from regulating Th1/Th17/Treg cell responses by activating PPAR-α/LXR-β signaling. These data suggest that fenofibrate may be a novel class of therapeutic agent for SS-associated dacryoadenitis.
Collapse
Affiliation(s)
- Xingyi Guo
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Weiyu Dang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Na Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Ying Wang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Deming Sun
- Doheny Eye Institute, And Department of Ophthalmology, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, California, United States
| | - Hong Nian
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Ruihua Wei
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| |
Collapse
|
16
|
Li S, Tang L, Zhou J, Anchouche S, Li D, Yang Y, Liu Z, Wu J, Hu J, Zhou Y, Yin J, Liu Z, Li W. Sleep deprivation induces corneal epithelial progenitor cell over-expansion through disruption of redox homeostasis in the tear film. Stem Cell Reports 2022; 17:1105-1119. [PMID: 35487212 PMCID: PMC9133657 DOI: 10.1016/j.stemcr.2022.03.017] [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: 09/27/2021] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 12/21/2022] Open
Abstract
Sleep deficiency, a common public health problem, causes ocular discomfort and affects ocular surface health. However, the underlying mechanism remains unclear. Herein, we identified that short-term sleep deprivation (SD) resulted in hyperproliferation of corneal epithelial progenitor cells (CEPCs) in mice. The expression levels of p63 and Keratin 14, the biomarkers of CEPCs, were upregulated in the corneal epithelium after short-term SD. In addition, SD led to elevated levels of reactive oxygen species (ROS), and subsequent decrease in antioxidant capacity, in the tear film. Exogenous hydrogen peroxide (H2O2) could directly stimulate the proliferation of CEPCs in vivo and in vitro. Topical treatment of antioxidant L-glutathione preserved the over-proliferation of CEPCs and attenuated corneal epithelial defects in SD mice. Moreover, the activation of the phosphoinositide 3-kinase (PI3K)/AKT signaling pathway is essential to ROS-stimulated cell proliferation in CEPCs. However, long-term SD ultimately led to early manifestation of limbal stem cell deficiency. Sleep deprivation induces the over-expansion of corneal epithelial progenitor cells (CEPCs) Sleep deprivation disrupts redox homeostasis in the tear film PI3K/AKT signaling pathway activation is essential to ROS-stimulated CEPC over-proliferation Topical L-glutathione treatment attenuates CEPC over-proliferation
Collapse
Affiliation(s)
- Sanming Li
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, Fujian 361104, China; Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Liying Tang
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, Fujian 361104, China; Department of Ophthalmology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361004, China
| | - Jing Zhou
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, Fujian 361104, China
| | - Sonia Anchouche
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts 02114, USA; Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, Ontario 027399, Canada
| | - Dian Li
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Yiran Yang
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, Fujian 361104, China
| | - Zhaolin Liu
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, Fujian 361104, China
| | - Jieli Wu
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, Fujian 361104, China
| | - Jiaoyue Hu
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, Fujian 361104, China
| | - Yueping Zhou
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, Fujian 361104, China
| | - Jia Yin
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Zuguo Liu
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, Fujian 361104, China; Department of Ophthalmology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361104, China; Xiamen University Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, Fujian 361004, China; Department of Ophthalmology, the First Affiliated Hospital of University South China, Hengyang, Hunan 421200, China.
| | - Wei Li
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, Fujian 361104, China; Department of Ophthalmology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361104, China; Xiamen University Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, Fujian 361004, China.
| |
Collapse
|
17
|
Wang S, He X, Li Q, Zhang Y, Hu J, Zong R, Zhuang J, Quantock AJ, Gao Y, Li W, Liu Z. Obstructive Sleep Apnea Affects Lacrimal Gland Function. Invest Ophthalmol Vis Sci 2022; 63:3. [PMID: 35238868 PMCID: PMC8899859 DOI: 10.1167/iovs.63.3.3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Purpose To determine the effect of obstructive sleep apnea syndrome (OSA) on lacrimal gland function and its mechanism. Methods Male mice aged seven to eight weeks were housed in cages with cyclic intermittent hypoxia to mimic OSA, and the control group was kept in a normal environment. Slit-lamp observation, fluorescein staining, and corneal sensitivity detection are used to assess cornea changes. Tear secretion was detected by phenol red cotton thread, and the pathological changes of lacrimal gland were observed by hematoxylin and eosin staining, oil red O staining, cholesterol and triglyceride kits, immunofluorescence staining, immunohistochemical staining, real-time polymerase chain reaction, transmission electron microscopy, and Western blot. Results Studies revealed a decreased tear secretion, corneal epithelial defects and corneal hypersensitivity. Myoepithelial cell damage, abnormal lipid accumulation, reduced cell proliferation, increased apoptosis and inflammatory cell infiltration in the lacrimal gland were also seen. Hifα and NF-κB signaling pathways, moreover, were activated, while Pparα was downregulated, in the lacrimal glands of OSA mice. Fenofibrate treatment significantly alleviated pathological changes of the lacrimal gland induced by OSA. Conclusion OSA disturbs the Hifα/Pparα/NF-κB signaling axis, which affects lacrimal gland structure and function and induces dry eye.
Collapse
Affiliation(s)
- Shaopan Wang
- Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.,Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen University, Xiamen, Fujian, China.,Institute of Artificial Intelligence, Xiamen University, Xiamen, Fujian, China
| | - Xin He
- Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.,Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen University, Xiamen, Fujian, China.,Department of Ophthalmology, the First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, China
| | - Qingmin Li
- Department of Ophthalmology, the Second Affiliated Hospital, Fujian Medical University, Quanzhou, Fujian, China
| | - Yuhan Zhang
- Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.,Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen University, Xiamen, Fujian, China
| | - Jiaoyue Hu
- Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.,Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen University, Xiamen, Fujian, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, China.,Xiamen University Affiliated Xiamen Eye Center, Xiamen University, Xiamen, Fujian, China
| | - Rongrong Zong
- Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.,Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen University, Xiamen, Fujian, China
| | - Jingyi Zhuang
- Department of Ophthalmology, the First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, China
| | - Andrew J Quantock
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, Wales, United Kingdom
| | - Yingying Gao
- Department of Ophthalmology, the Second Affiliated Hospital, Fujian Medical University, Quanzhou, Fujian, China
| | - Wei Li
- Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.,Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen University, Xiamen, Fujian, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, China.,Xiamen University Affiliated Xiamen Eye Center, Xiamen University, Xiamen, Fujian, China
| | - Zuguo Liu
- Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.,Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen University, Xiamen, Fujian, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, China.,Xiamen University Affiliated Xiamen Eye Center, Xiamen University, Xiamen, Fujian, China.,Institute of Artificial Intelligence, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
18
|
Almutairi R, Algezlan S, Bayamin R, Alrumaih S, Almutairi R, Alkahtani R, Almazrou AA. The Association Between Dry Eye and Sleep Quality Among the Adult Population of Saudi Arabia. Cureus 2022; 14:e22736. [PMID: 35386485 PMCID: PMC8969758 DOI: 10.7759/cureus.22736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2022] [Indexed: 11/30/2022] Open
Abstract
Background and objective Dry eye disease (DED) is one of the most prevalent ocular diseases worldwide. DED symptoms can result from disturbances to the homeostasis of the middle tear film layer (aqueous layer), including inflammation, pain, and eye discomfort, which can have a negative impact on individuals’ quality of life and daily activities. Sleep disorders are highly prevalent among patients with DED, and the incidence of sleep disturbances in DED patients has been reported to be as high as 40%. Decreased sleep quality can aggravate dry eye symptoms by increasing tear osmolarity and decreasing tear production. In this study, we aimed to investigate the association between DED and sleep quality in the adult population of Saudi Arabia. Methods This cross-sectional study was conducted among adult patients aged 18 years and above in Saudi Arabia in August 2021. A validated Arabic version of the Pittsburgh Sleep Quality Index (PSQI) was used to evaluate sleep quality, and the Ocular Surface Disease Index (OSDI) questionnaire was employed to diagnose DED. Data collection and analysis were performed using the SPSS Statistics software (IBM, Armonk, NY). Results A total of 234 subjects were analyzed, and 59.8% of the participants were women. Our tool suggested that 71.4% of the included participants had severe DED, 15% had moderate DED, and 13.7% had mild DED. However, 40.6% of the participants reported that they had not been diagnosed with DED previously and 34.6% had no previous DED symptoms. The mean total PSQI score was 8.63 ±2.23, with the highest score recorded for component 2: sleep latency (1.73) and the lowest score recorded for component 4: habitual sleep efficiency (0.20). Poor sleep quality as assessed by PSQI showed a significant positive correlation with the severity of DED as assessed by OSDI. Conclusion The significant positive correlation between poor sleep quality and DED indicated that patients with DED had a higher risk of poor sleep quality compared to healthy patients. Patients with DED should be educated about the steps and techniques to improve their sleep patterns.
Collapse
|
19
|
Zhu Y, Huang X, Lin L, Di M, Chen R, Fang F, Jin X. Sleep Quality Is Associated With Severe Meibomian Gland Disruption in Dry Eye. Front Med (Lausanne) 2022; 9:812705. [PMID: 35252250 PMCID: PMC8891226 DOI: 10.3389/fmed.2022.812705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/26/2022] [Indexed: 11/13/2022] Open
Abstract
PurposeTo investigate the association between sleep quality and meibomian gland dropout characteristics in dry eye patients.MethodsThis cross-sectional study involved 172 dry eye patients with no history of conditions or factors that could confound dry eye disease (DED) and/or meibomian gland dropout. Participants underwent a comprehensive anterior eye assessment. The validated Athens Insomnia Scale (AIS) and Pittsburgh Sleep Quality Index (PSQI) were used to assess sleep quality. The measured outcomes were dry eye symptoms via the Ocular Surface Disease Index (OSDI), tear breakup time (TBUT), corneal fluorescein staining, meibomian gland function, and extent of meibomian gland dropout.ResultsOf the dry eye participants, 34.9% had severe meibomian gland dropout (SMD) and 41.3% of the subjects had poor sleep quality. Patients with poor sleep quality had greater Meibomian gland dropout while the sleep AIS and PSQI scores were significantly correlated with Meibomian gland dropout (r = 0.495, p < 0.001; r = 0.24, p = 0.002; respectively). SMD patients had worse scores on all components of the PSQI (all p < 0.001, corrected for age and sex). Use of sleep medication, poor habitual sleep efficiency, and sleep disturbance were particularly prevalent in SMD patients as compared to Non-severe meibomian gland dropout (NSMD) patients. Multivariate logistic regression analysis revealed that sleep quality was eventually associated with female gender (p = 0.042), OSDI (p = 0.004), TBUT (p = 0.036), and Meibomian gland dropout score (p < 0.001).ConclusionIt was found that greater meibomian gland dropout in poor sleep quality individuals is especially related to use of sleep medication, poor habitual sleep efficiency, and sleep disturbance. This finding suggests a need for long-term studies of anterior eye health in people with poor sleep quality.
Collapse
Affiliation(s)
- Yirui Zhu
- Eye Center, Affiliated Second Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaodan Huang
- Eye Center, Affiliated Second Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lin Lin
- Eye Center, Affiliated Second Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Mengshu Di
- Eye Center, Affiliated Second Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ruida Chen
- Eye Center, Affiliated Second Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Eye Department, Affiliated Dongyang Hospital, Wenzhou Medical University, Dongyang, China
| | - Fei Fang
- Eye Center, Affiliated Second Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of ENT, Wuning County People's Hospital, Jiujiang, China
| | - Xiuming Jin
- Eye Center, Affiliated Second Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Xiuming Jin
| |
Collapse
|
20
|
Zheng Q, Li S, Wen F, Lin Z, Feng K, Sun Y, Bao J, Weng H, Shen P, Lin H, Chen W. The Association Between Sleep Disorders and Incidence of Dry Eye Disease in Ningbo: Data From an Integrated Health Care Network. Front Med (Lausanne) 2022; 9:832851. [PMID: 35187009 PMCID: PMC8854755 DOI: 10.3389/fmed.2022.832851] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 01/10/2022] [Indexed: 11/17/2022] Open
Abstract
PURPOSE To investigate the association between sleep disorders and dry eye disease (DED) in Ningbo, China. METHODS Our data came from the Yinzhou Health Information System (HIS), including 257932 patients and was based on a 1:1 matching method (sleep disorder patients vs. patients without sleep disorders) during 2013-2020. Sleep disorders and DED were identified using ICD-10 codes. Cox proportional hazards regression was used to identify the association between sleep disorders and DED. RESULTS The eight-year incidence of DED was significantly higher in participants with diagnosis of sleep disorders (sleep disorders: 50.66%, no sleep disorders: 16.48%, P < 0.01). Sleep disorders were positively associated with the diagnosis of DED (HR: 3.06, 95% CI: 2.99-3.13, P < 0.01), when sex, age, hypertension, diabetes and other systemic diseases were adjusted. In the sleep disorders patients, advancing age, female sex, and presence of coexisting disease (hypertension, diabetes, hyperlipidemia, thyroid disease, depression, heart disease, and arthritis) were significantly associated with the development of DED by the multivariate cox regression analysis (all P < 0.05).In addition, there was a significantly positive association between estazolam and the incidence of DED in both sleep disorder and non-sleep disorder groups (all P < 0.05). CONCLUSIONS Sleep disrder was associated with a three-time increased risk of DED. This association can be helpful in effective management of both sleep disorders and DED.
Collapse
Affiliation(s)
- Qinxiang Zheng
- The Affiliated Ningbo Eye Hospital of Wenzhou Medical University, Ningbo, China
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| | - Saiqing Li
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| | - Feng Wen
- The Affiliated Ningbo Eye Hospital of Wenzhou Medical University, Ningbo, China
| | - Zhong Lin
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| | - Kemi Feng
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| | - Yexiang Sun
- Department of Chronic Diseases and Health Promotion, Yinzhou District Center for Disease Control and Prevention, Ningbo, China
| | - Jie Bao
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| | - Hongfei Weng
- The Affiliated Ningbo Eye Hospital of Wenzhou Medical University, Ningbo, China
| | - Peng Shen
- Department of Chronic Diseases and Health Promotion, Yinzhou District Center for Disease Control and Prevention, Ningbo, China
| | - Hongbo Lin
- Department of Chronic Diseases and Health Promotion, Yinzhou District Center for Disease Control and Prevention, Ningbo, China
| | - Wei Chen
- The Affiliated Ningbo Eye Hospital of Wenzhou Medical University, Ningbo, China
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
21
|
Li A, Zhang X, Guo Y, Wang J, Hao Y, Gu Y, Jie Y. The Association Between Dry Eye and Sleep Disorders: The Evidence and Possible Mechanisms. Nat Sci Sleep 2022; 14:2203-2212. [PMID: 36545475 PMCID: PMC9762265 DOI: 10.2147/nss.s378751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 11/01/2022] [Indexed: 12/16/2022] Open
Abstract
Dry eye is a disease that severely affects patients' quality of life, increasing the global burden on public health and finance. There is growing evidence that a poor lifestyle is a significant risk factor for dry eye. Along with the development of society, sleep, as a way of life, is also constantly changing. The main manifestations of sleep disorders are reduced sleep time, circadian rhythm disturbances, and sleep breathing disturbances. Sleep disorders and their secondary systemic diseases have attracted wide attention in recent years. This review mainly explored the correlation between sleep disorders and dry eye, and found that sleep-related problems and other factors potentially leading from sleep disorders could be critical factors for dry eye. These results suggest that ophthalmologists should pay attention to the sleep health problems in patients with dry eye, and we hope that this paper can provide help for future research in this field.
Collapse
Affiliation(s)
- Ao Li
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Xiaozhao Zhang
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Yihan Guo
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Jingyi Wang
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Yiran Hao
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Yixuan Gu
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Ying Jie
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
22
|
Nguyen DD, Luo LJ, Lai JY. Thermogels containing sulfated hyaluronan as novel topical therapeutics for treatment of ocular surface inflammation. Mater Today Bio 2021; 13:100183. [PMID: 34927046 PMCID: PMC8649391 DOI: 10.1016/j.mtbio.2021.100183] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/02/2021] [Indexed: 12/26/2022] Open
Abstract
The development of long lasting therapeutic agents is critically important for efficient treatment of chronic diseases. We herein report a rational strategy to develop a therapeutic thermogel featured with prolonged anti-inflammatory and corneal-protective effects. Specifically, a hyaluronic acid with different sulfation degrees and an amine-terminated poly(N-isopropylacrylamide) are conjugated to achieve the thermogels. In vitro studies reveal that the thermogels are highly biocompatible to statens seruminstitut rabbit cornea cells and their anti-inflammatory properties are strongly dependent on the sulfation degree. In a rabbit model of ocular inflammation, single-dose topical administration of a thermogel formulation could repair defects in corneal epithelium (∼99% thickness restored), prevent corneal cell apoptosis (∼68.3% cells recovered), and suppress ocular surface inflammation (∼4-fold decrease) for a follow-up period of 7 days. This high treatment efficacy of the thermogel can be attributed to its potent inhibition in selectin-mediated leukocyte infiltration as well as effective corneal protection. These findings show a great promise for topical treatment of ocular inflammation and advancement of ophthalmic formulations using the bioactive thermogel as a therapeutic component that is not rapidly cleared from the eye and thus considerably reduces administration times. Sulfated hyaluronan thermogels served as intrinsic therapeutic agents. Thermogels exert inhibitory effects on selectin-mediated leukocyte infiltration. Sulfation degree is a key to achieve superior therapeutic thermogels. Highly sulfated agent reveals potent anti-inflammatory/corneal-protective effects. Single dose reduces corneal inflammation by 4-folds at 7 days post-instillation.
Collapse
Affiliation(s)
- Duc Dung Nguyen
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan, 33302, Taiwan
| | - Li-Jyuan Luo
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan, 33302, Taiwan
| | - Jui-Yang Lai
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan, 33302, Taiwan
- Department of Ophthalmology, Chang Gung Memorial Hospital, Linkou, Taoyuan, 33305, Taiwan
- Department of Materials Engineering, Ming Chi University of Technology, New Taipei City, 24301, Taiwan
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, 33303, Taiwan
- Corresponding author. Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan, 33302, Taiwan.
| |
Collapse
|
23
|
Wu J, Wang J, Wang L, Huang Y. Topical retinoic acid induces corneal strengthening by upregulating transglutaminase 2 in murine cornea. Exp Eye Res 2021; 214:108850. [PMID: 34861212 DOI: 10.1016/j.exer.2021.108850] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 09/22/2021] [Accepted: 11/15/2021] [Indexed: 01/23/2023]
Abstract
Transglutaminase 2 (TG2) is the most abundant crosslinking enzyme in murine and human cornea, while retinoids are well-known inducers of TG2 expression. This study aims to determine if the retinoic acid supplementation can increase corneal stiffness by crosslinking through upregulating the corneal TG2 expression. The right eyes of C57BL/6 mice were treated with 2 × 10-2M retinol palmitate (VApal) eyedrops or control eyedrops and hold for 30 min, once a day for 28 consecutive days. The WB and qPCR results showed increased expression of TG2 in murine cornea with the prolongation of VApal eyedrop application. After 28 days of VApal eyedrop treatment, the increased TG2 were found catalytically active and distributed in corneal epithelium and stroma as detected by 5-(biotinamido) pentylamine (5-BP) incorporation method and immunofluorescence staining. The transmission electron microscope image revealed that VApal treated cornea manifested with increased collagen density in anterior and middle layer of stroma. The higher elastic module was found among VApal treated cornea by nano-indentation test. In cultured corneal epithelial cells and keratocytes, all-trans retinoid acid (ATRA) treatment increased the content of TG2 in cell lysis and in culture medium. These results indicate that retinoic acid induce the reinforcement of the cornea by TG2 mediated crosslinking via increasing the TG2 expression in corneal epithelium and keratocyte. As TG2 was found to be less in the cornea of keratoconus patients in several RNA-sequencing studies, retinoic acid could serve as a non-invasive prevention method for keratoconus progression.
Collapse
Affiliation(s)
- Jie Wu
- Medical School of Chinese PLA, Beijing, 100089, China; Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, 100089, China
| | - Junyi Wang
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, 100089, China
| | - Liqiang Wang
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, 100089, China.
| | - Yifei Huang
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, 100089, China.
| |
Collapse
|
24
|
Lee D, Tomita Y, Allen W, Tsubota K, Negishi K, Kurihara T. PPARα Modulation-Based Therapy in Central Nervous System Diseases. Life (Basel) 2021; 11:life11111168. [PMID: 34833044 PMCID: PMC8622664 DOI: 10.3390/life11111168] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 10/29/2021] [Accepted: 10/30/2021] [Indexed: 12/11/2022] Open
Abstract
The burden of neurodegenerative diseases in the central nervous system (CNS) is increasing globally. There are various risk factors for the development and progression of CNS diseases, such as inflammatory responses and metabolic derangements. Thus, curing CNS diseases requires the modulation of damaging signaling pathways through a multitude of mechanisms. Peroxisome proliferator-activated receptors (PPARs) are a family of nuclear hormone receptors (PPARα, PPARβ/δ, and PPARγ), and they work as master sensors and modulators of cellular metabolism. In this regard, PPARs have recently been suggested as promising therapeutic targets for suppressing the development of CNS diseases and their progressions. While the therapeutic role of PPARγ modulation in CNS diseases has been well reviewed, the role of PPARα modulation in these diseases has not been comprehensively summarized. The current review focuses on the therapeutic roles of PPARα modulation in CNS diseases, including those affecting the brain, spinal cord, and eye, with recent advances. Our review will enable more comprehensive therapeutic approaches to modulate PPARα for the prevention of and protection from various CNS diseases.
Collapse
Affiliation(s)
- Deokho Lee
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo 160-8582, Japan;
- Department of Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan;
| | - Yohei Tomita
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo 160-8582, Japan;
- Department of Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan;
- Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
- Correspondence: (Y.T.); (T.K.); Tel.: +1-617-919-2533 (Y.T.); +81-3-5636-3204 (T.K.)
| | - William Allen
- Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | | | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan;
| | - Toshihide Kurihara
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo 160-8582, Japan;
- Department of Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan;
- Correspondence: (Y.T.); (T.K.); Tel.: +1-617-919-2533 (Y.T.); +81-3-5636-3204 (T.K.)
| |
Collapse
|
25
|
Jin M, Wang Y, An X, Kang H, Wang Y, Wang G, Gao Y, Wu S, Reinach PS, Liu Z, Xue Y, Li C. Phenotypic and transcriptomic changes in the corneal epithelium following exposure to cigarette smoke. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 287:117540. [PMID: 34147784 DOI: 10.1016/j.envpol.2021.117540] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 06/12/2023]
Abstract
Cigarette smoke extract (CSE), a complex mixture of compounds, contributes to a range of eye diseases; however, the underlying pathophysiological responses to tobacco smoke remain ambiguous. The purpose of the present study was to evaluate the cigarette smoke-induced phenotypic and transcriptomic changes in the corneal epithelium with a view to elucidating the likely underlying mechanism. Accordingly, for the first time, we characterized the genome-wide effects of CSE on the corneal epithelium. The ocular surface of the mice in the experimental groups was exposed to CSE for 1 h per day for a period of one week, while mice in the control group were exposed to preservative-free artificial tears. Corneal fluorescein staining, in vivo confocal microscopy and scanning electron microscopy were performed to examine the corneal ultrastructure. Transcriptome sequencing and bioinformatics analysis were performed followed by RT-qPCR to validate gene expression changes. The results indicate that CSE exposure disrupted the structural integrity of the superficial epithelium, decreased the density of microvilli, and compromised the corneal epithelial barrier intactness. RNA-seq revealed 667 differentially expressed genes, and functional analysis highlighted the enhancement of several biological processes such as antioxidant activity and the response to oxidative stress. Moreover, the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis showed that glutathione metabolism and drug metabolism cytochrome P450 were the most relevant pathways contributing to the effects of CSE on the corneal epithelium. Protein-protein interaction (PPI) network analysis illustrated that GCLC, NQO1, and HMOX1 were the most relevant nodes. In conclusion, the present study indicates that CSE exposure induces changes in the phenotype and genotype of the corneal epithelium. The antioxidant response element is essential for counteracting the effects of cigarette smoke on this tissue layer. These results shed novel insights into how cigarette smoke damages this ocular surface.
Collapse
Affiliation(s)
- Mengyi Jin
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Yanzi Wang
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Xiaoya An
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, 361102, China; School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Honghua Kang
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Yixin Wang
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, 361102, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Guoliang Wang
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, 361102, China; School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Yang Gao
- College of Environment and Ecology, Xiamen University, Xiamen, 361102, China
| | - Shuiping Wu
- College of Environment and Ecology, Xiamen University, Xiamen, 361102, China
| | - Peter S Reinach
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Zuguo Liu
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, 361102, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Yuhua Xue
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Cheng Li
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, 361102, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, 361102, China.
| |
Collapse
|
26
|
Wu J, Wu T, Zheng S, Huang Y, Wang L. Low-dose repeated exposure to chemical surfactant impairs corneal epithelium: When personal cleaning products entering the eye. Exp Eye Res 2021; 210:108696. [PMID: 34228968 DOI: 10.1016/j.exer.2021.108696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 06/17/2021] [Accepted: 06/30/2021] [Indexed: 02/03/2023]
Abstract
Studies have reported that the incidence of ocular discomfort in people who often wear makeup is higher than that in the normal population. The incidence of ocular discomfort of these people may be also related to the daily ocular exposure to chemical surfactants during cleaning. The objectives of this study were to explore morphological and pathological changes in the murine ocular surface after low-dose repeated exposure to disodium cocoamphodiacetate (DC), a kind of chemical surfactant widely used in personal cleaning products, and to investigate the possible mechanisms. DC was administered in low dose (0.1%) to the ocular surface of C56BL/6 once daily for two weeks. We found that there were an increase of sodium fluorescein staining on the cornea, a significant thinning of corneal epithelial thickness, and increased TUNEL-positive cells in corneal epithelium in vivo. DC treatment also modulated the distribution of K14+ and P63+ epithelia from the limbal to the center on the cornea. In cultured murine corneal epithelial progenitor cell line (TKE2), DC treatment induced cell detachment and decreased the activation of Ak strain transforming protein (AKT), and extracellular signal-regulated kinase (ERK). And DC increased TUNEL-positive cells in vitro with increased expression of cleaved Caspase3 and B-cell lymphoma-2 associated X protein (Bax). Our results indicated that repeated low-dose DC exposure on ocular surface caused significant impairment on the structure and viability of the corneal epithelium by inhibiting epithelial proliferation and inducing apoptosis. It provides the foundations to understand the harmful effects of cleaning products daily exposure on the ocular surface.
Collapse
Affiliation(s)
- Jie Wu
- Medical School of Chinese PLA, Beijing, 100089, China; Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, 100089, China
| | - Tengyun Wu
- Medical School of Chinese PLA, Beijing, 100089, China; Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, 100089, China
| | - Shuo Zheng
- Medical School of Chinese PLA, Beijing, 100089, China; Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, 100089, China
| | - Yifei Huang
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, 100089, China
| | - Liqiang Wang
- Department of Ophthalmology, The Third Medical Center, Chinese PLA General Hospital, Beijing, 100089, China.
| |
Collapse
|
27
|
Escandon P, Vasini B, Whelchel AE, Nicholas SE, Matlock HG, Ma JX, Karamichos D. The role of peroxisome proliferator-activated receptors in healthy and diseased eyes. Exp Eye Res 2021; 208:108617. [PMID: 34010603 PMCID: PMC8594540 DOI: 10.1016/j.exer.2021.108617] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 12/23/2022]
Abstract
Peroxisome Proliferator-Activated Receptors (PPARs) are a family of nuclear receptors that play essential roles in modulating cell differentiation, inflammation, and metabolism. Three subtypes of PPARs are known: PPAR-alpha (PPARα), PPAR-gamma (PPARγ), and PPAR-beta/delta (PPARβ/δ). PPARα activation reduces lipid levels and regulates energy homeostasis, activation of PPARγ results in regulation of adipogenesis, and PPARβ/δ activation increases fatty acid metabolism and lipolysis. PPARs are linked to various diseases, including but not limited to diabetes, non-alcoholic fatty liver disease, glaucoma and atherosclerosis. In the past decade, numerous studies have assessed the functional properties of PPARs in the eye and key PPAR mechanisms have been discovered, particularly regarding the retina and cornea. PPARγ and PPARα are well established in their functions in ocular homeostasis regarding neuroprotection, neovascularization, and inflammation, whereas PPARβ/δ isoform function remains understudied. Naturally, studies on PPAR agonists and antagonists, associated with ocular pathology, have also gained traction with the development of PPAR synthetic ligands. Studies on PPARs has significantly influenced novel therapeutics for diabetic eye disease, ocular neuropathy, dry eye, and age-related macular degeneration (AMD). In this review, therapeutic potentials and implications will be highlighted, as well as reported adverse effects. Further investigations are necessary before any of the PPARs ligands can be utilized, in the clinics, to treat eye diseases. Future research on the prominent role of PPARs will help unravel the complex mechanisms involved in order to prevent and treat ocular diseases.
Collapse
Affiliation(s)
- Paulina Escandon
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Brenda Vasini
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Amy E Whelchel
- Department of Physiology, University of Oklahoma Health Sciences Center, 940 Stanton L Young, Oklahoma City, OK, USA
| | - Sarah E Nicholas
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - H Greg Matlock
- Department of Physiology, University of Oklahoma Health Sciences Center, 940 Stanton L Young, Oklahoma City, OK, USA
| | - Jian-Xing Ma
- Department of Physiology, University of Oklahoma Health Sciences Center, 940 Stanton L Young, Oklahoma City, OK, USA; Harold Hamm Oklahoma Diabetes Center, 1000 N Lincoln Blvd, Oklahoma City, OK, USA
| | - Dimitrios Karamichos
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA.
| |
Collapse
|
28
|
Ji S, Mao X, Zhang Y, Ye L, Dai J. Contribution of M-opsin-based color vision to refractive development in mice. Exp Eye Res 2021; 209:108669. [PMID: 34126082 DOI: 10.1016/j.exer.2021.108669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 06/02/2021] [Accepted: 06/08/2021] [Indexed: 11/26/2022]
Abstract
M-opsin, encoded by opn1mw gene, is involved in green-light perception of mice. The role of M-opsin in emmetropization of mice remains uncertain. To answer the above question, 4-week-old wild-type (WT) mice were exposed to white light or green light (460-600 nm, a peak at 510 nm) for 12 weeks. Refractive development was estimated biweekly. After treatment, retinal function was assessed using electroretinogram (ERG). Dopamine (DA) in the retina was evaluated by high-performance liquid chromatography, M-opsin and S-opsin protein levels by Western blot and ELISA, and mRNA expressions of opn1mw and opn1sw by RT-PCR. Effects of M-opsin were further verified in Opn1mw-/- and WT mice raised in white light for 4 weeks. Refractive development was examined at 4, 6, and 8 weeks after birth. The retinal structure was estimated through hematoxylin and eosin staining (H&E) and transmission electron microscopy (TEM). Retinal wholemounts from WT and Opn1mw-/- mice were co-immunolabeled with M-opsin and S-opsin, their distribution and quantity were then assayed by immunofluorescence staining (IF). Expression of S-opsin protein and opn1sw mRNA were determined by Western blot, ELISA, or RT-PCR. Retinal function and DA content were analyzed by ERG and liquid chromatography tandem-mass spectrometry (LC-MS/MS), respectively. Lastly, visual cliff test was used to evaluate the depth perception of the Opn1mw-/- mice. We found that green light-treated WT mice were more myopic with increased M-opsin expression and decreased DA content than white light-treated WT mice after 12-week illumination. No electrophysiologic abnormalities were recorded in mice exposed to green light compared to those exposed to white light. A more hyperopic shift was further observed in 8-week-old Opn1mw-/- mice in white light with lower DA level and weakened cone function than the WT mice under white light. Neither obvious structural disruption of the retina nor abnormal depth perception was found in Opn1mw-/- mice. Together, these results suggested that the M-opsin-based color vision participated in the refractive development of mice. Overexposure to green light caused myopia, but less perception of the middle-wavelength components in white light promoted hyperopia in mice. Furthermore, possible dopaminergic signaling pathway was suggested in myopia induced by green light.
Collapse
Affiliation(s)
- Shunmei Ji
- Department of Ophthalmology, Eye & ENT Hospital Affiliated to Fudan University, Shanghai, China; Department of Ophthalmology, Zhongshan Hospital Affiliated to Fudan University, Shanghai, China; NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences (Fudan University), Shanghai, China
| | - Xiuyu Mao
- Department of Ophthalmology, Eye & ENT Hospital Affiliated to Fudan University, Shanghai, China; NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences (Fudan University), Shanghai, China
| | - Yifan Zhang
- Department of Ophthalmology, Eye & ENT Hospital Affiliated to Fudan University, Shanghai, China; NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences (Fudan University), Shanghai, China
| | - Lin Ye
- Department of Ophthalmology, Eye & ENT Hospital Affiliated to Fudan University, Shanghai, China; NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences (Fudan University), Shanghai, China
| | - Jinhui Dai
- Department of Ophthalmology, Eye & ENT Hospital Affiliated to Fudan University, Shanghai, China; Department of Ophthalmology, Zhongshan Hospital Affiliated to Fudan University, Shanghai, China.
| |
Collapse
|
29
|
PPARα Agonist Oral Therapy in Diabetic Retinopathy. Biomedicines 2020; 8:biomedicines8100433. [PMID: 33086679 PMCID: PMC7589723 DOI: 10.3390/biomedicines8100433] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/13/2020] [Accepted: 10/15/2020] [Indexed: 12/12/2022] Open
Abstract
Diabetic retinopathy (DR) is an eye condition that develops after chronically poorly-managed diabetes, and is presently the main cause for blindness on a global scale. Current treatments for DR such as laser photocoagulation, topical injection of corticosteroids, intravitreal injection of anti-vascular endothelial growth factor (VEGF) agents and vitreoretinal surgery are only applicable at the late stages of DR and there are possibilities of significant adverse effects. Moreover, the forms of treatment available for DR are highly invasive to the eyes. Safer and more effective pharmacological treatments are required for DR treatment, in particular at an early stage. In this review, we cover recently investigated promising oral pharmacotherapies, the methods of which are safer, easier to use, patient-friendly and pain-free, in clinical studies. We especially focus on peroxisome proliferator-activator receptor alpha (PPARα) agonists in which experimental evidence suggests PPARα activation may be closely related to the attenuation of vascular damages, including lipid-induced toxicity, inflammation, an excess of free radical generation, endothelial dysfunction and angiogenesis. Furthermore, oral administration of selective peroxisome proliferator-activated receptor alpha modulator (SPPARMα) agonists may induce hepatic fibroblast growth factor 21 expression, indirectly resulting in retinal protection in animal studies. Our review will enable more comprehensive approaches for understanding protective roles of PPARα for the prevention of DR development.
Collapse
|
30
|
He X, Zhao Z, Wang S, Kang J, Zhang M, Bu J, Cai X, Jia C, Li Y, Li K, Reinach PS, Quantock AJ, Liu Z, Li W. High-Fat Diet-Induced Functional and Pathologic Changes in Lacrimal Gland. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:2387-2402. [PMID: 32919976 DOI: 10.1016/j.ajpath.2020.09.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 08/30/2020] [Accepted: 09/03/2020] [Indexed: 12/31/2022]
Abstract
The lacrimal gland is critical for maintaining the homeostasis of the ocular surface microenvironment through secreting aqueous tears in mammals. Many systemic diseases such as Sjögren syndrome, rheumatoid arthritis, and diabetes can alter the lacrimal gland function, eventually resulting in aqueous tear-deficient dry eye. Here, a high-fat diet (HFD) experimental mouse model was used to clarify how hyperlipidemia affects lacrimal gland function. Aqueous tear secretion fell about 50% after 1 month on a HFD. Lipid droplets accumulated in the matrix and acinar cells of the lacrimal gland after this period, along with changes in the lipid metabolism, changes in gene expression levels, and disruption of fatty acid oxidative activity. Immune cell infiltration and rises in the gene expression levels of the inflammation-related cytokines Il1β, Tnfα, Tsg6, Il10, Mmp2, and Mmp9 were found. HFD also induced mitochondrial hypermegasoma, increased apoptosis, and decreased lacrimal gland acinar cell proliferation. Replacement of the HFD with the standard diet partially reversed pathologic changes in the lacrimal gland. Similarly, supplementing the HFD with fenofibrate also partially reversed the inhibited tear secretion and reduced lipid accumulation, inflammation, and oxidative stress levels. The authors conclude that a HFD induces pathophysiological changes and functional decompensation of the lacrimal gland. Therefore, ingestion of a HFD may be a causative factor of dry eye disease.
Collapse
Affiliation(s)
- Xin He
- Eye Institute of Xiamen University, Xiamen, China; School of Medicine, Xiamen University, Xiamen, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen University, Xiamen, China
| | - Zhongyang Zhao
- Eye Institute of Xiamen University, Xiamen, China; School of Medicine, Xiamen University, Xiamen, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen University, Xiamen, China
| | - Shaopan Wang
- Eye Institute of Xiamen University, Xiamen, China; School of Medicine, Xiamen University, Xiamen, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen University, Xiamen, China
| | - Jie Kang
- Eye Institute of Xiamen University, Xiamen, China; School of Medicine, Xiamen University, Xiamen, China; Xiang'an Hospital of Xiamen University, Xiamen, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen University, Xiamen, China
| | - Minjie Zhang
- Eye Institute of Xiamen University, Xiamen, China; School of Medicine, Xiamen University, Xiamen, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen University, Xiamen, China
| | - Jinghua Bu
- Eye Institute of Xiamen University, Xiamen, China; School of Medicine, Xiamen University, Xiamen, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen University, Xiamen, China
| | - Xiaoxin Cai
- Eye Institute of Xiamen University, Xiamen, China; School of Medicine, Xiamen University, Xiamen, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen University, Xiamen, China
| | - Changkai Jia
- Eye Institute of Xiamen University, Xiamen, China; School of Medicine, Xiamen University, Xiamen, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen University, Xiamen, China
| | - Yixuan Li
- School of Medicine, Xiamen University, Xiamen, China
| | | | | | - Andrew J Quantock
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, United Kingdom
| | - Zuguo Liu
- Eye Institute of Xiamen University, Xiamen, China; School of Medicine, Xiamen University, Xiamen, China; Xiang'an Hospital of Xiamen University, Xiamen, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen University, Xiamen, China; Xiamen University Affiliated Xiamen Eye Center, Xiamen, China.
| | - Wei Li
- Eye Institute of Xiamen University, Xiamen, China; School of Medicine, Xiamen University, Xiamen, China; Xiang'an Hospital of Xiamen University, Xiamen, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen University, Xiamen, China; Xiamen University Affiliated Xiamen Eye Center, Xiamen, China.
| |
Collapse
|
31
|
He H, Liang M, Li L, Luo S, Fang X, He H, Xiao X, Wu H, Lin Z. PPAR-α Agonist Fenofibrate Suppressed the Formation of Ocular Surface Squamous Metaplasia Induced by Topical Benzalkonium Chloride. Invest Ophthalmol Vis Sci 2020; 61:54. [PMID: 32232349 PMCID: PMC7401654 DOI: 10.1167/iovs.61.3.54] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Purpose To investigate the effects and mechanisms of the peroxisome proliferator-activated receptor alpha (PPAR-α) agonist fenofibrate on the formation of ocular surface squamous metaplasia induced by topical benzalkonium chloride (BAC) in a mouse model. Methods Ocular surface squamous metaplasia was induced in 16 days by topical BAC application in mice. During the period of induction, mice were divided into four groups: no additional treatment (BAC+UT), topical vehicle (BAC+Vehicle), topical fenofibrate (BAC+Feno), or topical fenofibrate plus intraperitoneal injection of MK886 (BAC+Feno+MK886). The parameters of tear film were evaluated on day 16, and eye specimens were collected. Histologic investigation; PAS assays; immunostaining for cytokeratin 10 (K10), Ki67, and F4/80; and PCR assays for TNF-α and IL-6 were performed. Cell Counting Kit 8 (CCK-8) assays were performed to evaluate the inhibitory effects of fenofibrate on RAW264.7 cells. Results Fenofibrate suppressed the formation of BAC-induced instable tear film. In the BAC+Feno group, the expression of K10 and Ki67 was lower than in the other three groups. The number of goblet cells was reduced in eyes of the BAC+UT and BAC+Vehicle groups but was maintained in eyes of the BAC+Feno group. The number of F4/80-positive cells and the levels of TNF-α and IL-6 mRNA were significantly reduced in the cornea of the BAC+Feno group. These effects of fenofibrate could be attenuated by MK886. The cell viability of RAW264.7 cells could be significantly inhibited by fenofibrate in a dose-dependent pattern. Conclusions Topical application of fenofibrate suppressed the formation of ocular surface squamous metaplasia, which might be mediated through the PPAR-α signaling pathway.
Collapse
|
32
|
Zhang C, Zheng Y, Li M, Zhang Z, Chang L, Ai M, Wang J, Zhao S, Li C, Zhou Z. Carboxymethyl Cellulose-Coated Tacrolimus Nonspherical Microcrystals for Improved Therapeutic Efficacy of Dry Eye. Macromol Biosci 2020; 20:e2000079. [PMID: 32537876 DOI: 10.1002/mabi.202000079] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/28/2020] [Indexed: 01/08/2023]
Abstract
Dry eye (DE) is a highly prevalent ocular surface disease which affects the quality of life and results in low working efficiency. Frequent instillation is required due to low bioavailability of conventional eye drops. The aim of this study is to develop a novel formulation of tacrolimus (TAC), routinely prescribed for DE, by combination of the microcrystal technology and layer-by-layer assembly. First, nonspherical tacrolimus microcrystals (TAC MCs) are synthesized by antisolvent-induced precipitation. These TAC MCs are modified by alternate deposition of poly(allylamine hydrochloride) (PAH) and carboxymethyl cellulose (CMC) subsequently to obtain CMC-coated TAC MCs (TAC-(PAH/CMC)3 ). The resultant formulations are evaluated in vivo in a mouse DE model induced by an intelligently controlled environmental system. Compared with commercially available TAC eye drops and the TAC MCs counterpart, TAC-(PAH/CMC)3 exhibits superior therapeutic performance with reduced drug instillation frequency, which is attributed to the nonspherical geometry of MCs, the lubricant, mucoadhesive effect of CMC, and the anti-inflammatory function of TAC. Therefore, TAC-(PAH/CMC)3 represents a better option for the management of DE.
Collapse
Affiliation(s)
- Caijie Zhang
- Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | | | - Min Li
- Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Biomedical Materials, Tianjin, 300192, China
| | - Zhongfang Zhang
- Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Lianqing Chang
- Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Mingyue Ai
- Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Biomedical Materials, Tianjin, 300192, China
| | - Jingjie Wang
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325027, China
| | - Shaozhen Zhao
- Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Chen Li
- Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Biomedical Materials, Tianjin, 300192, China
| | - Zhimin Zhou
- Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Biomedical Materials, Tianjin, 300192, China
| |
Collapse
|
33
|
Luo LJ, Nguyen DD, Lai JY. Long-acting mucoadhesive thermogels for improving topical treatments of dry eye disease. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 115:111095. [PMID: 32600699 DOI: 10.1016/j.msec.2020.111095] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 04/12/2020] [Accepted: 05/12/2020] [Indexed: 12/25/2022]
Abstract
Dry eye disease (DED) is the most common ocular disorder that causes persistent discomfort and blurry vision in patients. Despite pharmacotherapy strategies, the current topical administration of eye drops remains a great challenge owing to their low bioavailability and short residence time. Herein, we demonstrate an effective topical treatment of DED via rational design of a long-acting and mucoadhesive drug delivery system. Specifically, the drug carrier is a chemically ternary material system consisting of gelatin that serves as an enzyme-mediated degradable matrix, poly(N-isopropylacrylamide) as a thermo-responsive regulator, and lectin Helix pomatia agglutinin as a mucus-binding component. The long-acting drug release performance is exploited via initiator effects during the synthesis of the thermo-responsive polymer, while the mucoadhesive feature is inherited from the mucus-binding material. In a rabbit model of DED, a pharmacotherapy based on one-time topical administration of epigallocatechin gallate-loaded carrier onto the cul-de-sac could effectively repair the defective corneal epithelium via mitigating cellular inflammation, oxidative stress, and cell apoptosis for a sustained period over 14 days. These findings on the initiator and synergy effects in the development of the advanced ophthalmic formulation show great promise for efficient management of complex ocular diseases by a simple topical administration route.
Collapse
Affiliation(s)
- Li-Jyuan Luo
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan 33302, Taiwan, ROC
| | - Duc Dung Nguyen
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan 33302, Taiwan, ROC
| | - Jui-Yang Lai
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan 33302, Taiwan, ROC; Department of Ophthalmology, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan, ROC; Department of Materials Engineering, Ming Chi University of Technology, New Taipei City 24301, Taiwan, ROC.
| |
Collapse
|
34
|
Jiao X, Lu D, Pei X, Qi D, Huang S, Song Z, Gu J, Li Z. Type 1 diabetes mellitus impairs diurnal oscillations in murine extraorbital lacrimal glands. Ocul Surf 2020; 18:438-452. [PMID: 32360784 DOI: 10.1016/j.jtos.2020.04.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 04/20/2020] [Accepted: 04/23/2020] [Indexed: 01/09/2023]
Abstract
PURPOSE People with diabetes are at high risk of lacrimal gland dysfunction, but the underlying mechanism is not well understood. In this study, we determined how type 1 diabetes mellitus (T1DM) influences circadian homeostasis of the murine extraorbital lacrimal glands (ELGs). METHODS A T1DM animal model was established by systemic streptozotocin injection in C57BL/6J mice. After 5 weeks, ELGs were collected at 3-h intervals over a 24-h circadian cycle. Total extracted RNA was subjected to high-throughput RNA sequencing, and rhythmic transcriptional data were evaluated using the Jonckheere-Terpstra-Kendall algorithm, Kyoto Encyclopedia of Genes and Genomes pathway analysis, Phase Set Enrichment Analysis, and time series cluster analysis to determine the phase, rhythmicity, and unique signature of the transcripts over temporally coordinated expression. Additionally, mass, cell size, histology, and tear secretion of the ELGs were evaluated. RESULTS T1DM globally altered the composition of the ELG transcriptome. Specifically, T1DM significantly reprogrammed the circadian transcriptomic profiles of normal ELGs and reorganized core clock machinery. Unique temporal and clustering enrichment pathways were also rewired by T1DM. Finally, normal daily rhythms of mass, cell size, and tear secretion of mouse ELGs were significantly impaired by streptozotocin-induced diabetes. CONCLUSIONS T1DM significantly reprograms the diurnal oscillations of the lacrimal glands and impairs their structure and tear secretion. This information may reveal potential targets for improving lacrimal gland dysfunction in patients with diabetes.
Collapse
Affiliation(s)
- Xinwei Jiao
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Dingli Lu
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Xiaoting Pei
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Di Qi
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Shenzhen Huang
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Zongming Song
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Jianqin Gu
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Zhijie Li
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China.
| |
Collapse
|
35
|
The effect of OSAS risk, excessive daytime sleepiness, insomnia severity and sleep quality on dry eye syndrome. Sleep Biol Rhythms 2020. [DOI: 10.1007/s41105-020-00267-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
36
|
Wang X, Tang L, Zhang Z, Li W, Chen Y. Keratocytes promote corneal neovascularization through VEGFr3 induced by PPARα-inhibition. Exp Eye Res 2020; 193:107982. [PMID: 32092288 DOI: 10.1016/j.exer.2020.107982] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/30/2020] [Accepted: 02/19/2020] [Indexed: 12/11/2022]
Abstract
As the peroxisome proliferator - activated receptor alpha (PPARα) agonist, fenofibrate has been widely used to be a good lipid-regulating drug in the clinical application. In this study, we investigated the mechanism by which keratocytes inhibit the corneal neovascularization (CNV) through PPARα - activation. To do this, the CNV model was established by alkali burn, followed by being divided into three groups including control, fenofibrate and vehicle group. The expression of VEGFr3, MMP13 and PPARα in corneas of normal mouse and alkali-burned mouse was determined via quantitative RT- PCR (qRT-PCR) and Western blot analysis (WB). The CNV area was observed under a slit lamp microscope. The location of PPARα expression in the corneas was determined via immunohistochemistry. In cultured primary keratocytes, the effect of fenofibrate on PPARα, VEGFr3 and MMP13 expression was determined by qRT-PCR and WB. Besides, PPARα knockout (PPARα-/-) mouse CNV and keratocytes model were established to further confirm the effect of PPARα on VEGFr3 and MMP13 expression. We found that PPARα was expressed in epithelium, stroma and endothelium of the normal cornea, however, with relatively low level in the corneal stroma. Meanwhile, its expression was decreased markedly in the cornea during the stage of CNV formation. After treatment of fenofibrate, PPARα expression was promoted and the expression of VEGFr3 and MMP13 was inhibited in both CNV mice model and primary keratocytes, and CNV areas were decreased in CNV mice model. However, the results in PPARα-/- CNV and keratocytes model were opposite. Our results suggest that keratocytes could promote the expression of VEGFr3 and MMP13, and CNV formation through PPARα downregulation.
Collapse
Affiliation(s)
- Xue Wang
- Aier School of Ophthalmology, Central South University, Changsha, 410015, China; Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Medical College, Xiamen University, Xiamen, Fujian, China
| | - Liying Tang
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Medical College, Xiamen University, Xiamen, Fujian, China
| | - Zhaoqiang Zhang
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Medical College, Xiamen University, Xiamen, Fujian, China
| | - Wensheng Li
- Aier School of Ophthalmology, Central South University, Changsha, 410015, China; Shanghai Aier Eye Hospital, Shanghai, 200336, China.
| | - Yongxiong Chen
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Medical College, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
37
|
Chen Q, Ji C, Zheng R, Yang L, Ren J, Li Y, Han Y, Zhou P, Liu Z, Qiu Y. N-Palmitoylethanolamine Maintains Local Lipid Homeostasis to Relieve Sleep Deprivation-Induced Dry Eye Syndrome. Front Pharmacol 2020; 10:1622. [PMID: 32047441 PMCID: PMC6997544 DOI: 10.3389/fphar.2019.01622] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 12/13/2019] [Indexed: 11/13/2022] Open
Abstract
Sleep loss is a key factor associated with dry eye. Use of a “stick over water” mouse model revealed that sleep deprivation induces accumulation of lipids, hypertrophy, and dysfunction of the lacrimal gland. These changes result in decreased tear production and dry eye clinical signs. The specific pathophysiological mechanisms that contribute to dry eye remain unclear. In this study, we found that sleep deprivation decreased endogenous lipid palmitoylethanolamide (PEA) expression in the lacrimal gland. The reduced expression was mainly attributed to the decreased expression of N-acylated phosphatidylethanolamine–phospholipase D, the synthetic enzyme of PEA. Exogenous PEA treatment restored local lipid metabolism homeostasis in the lacrimal gland. This change was accompanied by reduced lipid deposition, maintenance of the endoplasmic reticulum and mitochondrial morphology, and improved acinar cell secretory function. PEA treatment also prevented damage to corneal barrier function and improved the dry eye clinical signs caused by sleep deprivation. The nuclear receptor peroxisome proliferator-activated receptor-α (PPAR-α) was found to mediate the PEA-associated improvements. We describe here for the first time that PEA is involved in sleep deprivation–induced lacrimal gland pathogenesis and dry eye development. PEA and its metabolizing enzymes may serve as adjunctive therapeutic targets for treatment of dry eye.
Collapse
Affiliation(s)
- Qi Chen
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.,Xiamen University Affiliated Xiamen Eye Center, Xiamen, China
| | - Chunyan Ji
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Ruihe Zheng
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Longhe Yang
- Engineering Research Center of Marine Biological Resource Comprehensive Utilization, Third Institute of Oceanography, State Oceanic Administration, Xiamen, China
| | - Jie Ren
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Yitian Li
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Yun Han
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.,Xiamen University Affiliated Xiamen Eye Center, Xiamen, China
| | - Pan Zhou
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Zuguo Liu
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.,Xiamen University Affiliated Xiamen Eye Center, Xiamen, China
| | - Yan Qiu
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, China.,Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, China.,Xiamen University Affiliated Xiamen Eye Center, Xiamen, China
| |
Collapse
|
38
|
Chen SY, Xie C, Zhu H, Shen Y. Effects of epidermal growth factor on transforming growth factor-beta1-induced epithelial-mesenchymal transition and potential mechanism in human corneal epithelial cells. Int J Ophthalmol 2020; 13:11-20. [PMID: 31956565 DOI: 10.18240/ijo.2020.01.03] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 11/17/2019] [Indexed: 01/09/2023] Open
Abstract
AIM To evaluate the effects of epidermal growth factor (EGF) on transforming growth factor-beta1 (TGF-β1)-induced epithelial-mesenchymal transition (EMT) in human corneal epithelial cells (HCECs). METHODS HCECs were cultured and treated with TGF-β1 for establishing the model of EMT in vitro. Biological effect of EGF on TGF-β1-induced EMT was evaluated. Proteins and mRNAs expression changes of E-cadherin, N-cadherin and Fibronectin (EMT-relative markers) after TGF-β1 or TGF-β1 combined EGF treatment were detected by Western blot and RT-PCR, respectively. Viability and migration of HCECs were measured by CCK-8, transwell cell migration assay and cell scratch wound healing assay. Activation of Smad2, ERK, p38, JNK and Akt signaling pathways were evaluated by Western blot. Inhibitors of relevant signaling pathways were added to the HCECs to explore the key signal mechanism. RESULTS With treatment of TGF-β1 only, three EMT-relative proteins and mRNA expression showed that EMT up-regulated in a concentration-dependent and time-dependent manner, with significantly decreasing cell viability (TGF-β1≥5 ng/mL, P<0.05) and increasing cell migration (TGF-β1≥5 ng/mL, P<0.01). The phosphorylation of Smad2 and p38 was a key process of TGF-β1-induced EMT. Meanwhile, EMT-relative proteins and mRNA expression showed that EGF inhibited TGF-β1-indued EMT, with significantly increasing cell viability (EGF≥10 ng/mL, P<0.01). It was noteworthy that EGF significantly enhanced cell migration although EMT was inhibited (EGF≥10 ng/mL, P<0.01), and the blockage of p38 (by SB202190, a p38 inhibitor) was a potential mechanism of this phenomenon. CONCLUSION EGF inhibits TGF-β1-induced EMT via suppressive p38, and promotes cells proliferation and migration in a non-EMT process by inhibiting p38 pathway.
Collapse
Affiliation(s)
- Shu-Yang Chen
- Department of Ophthalmology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China.,Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang Province, China
| | - Chen Xie
- Department of Ophthalmology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China.,Clinical Research Center, the First Affiliate Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Hong Zhu
- Department of Ophthalmology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China.,Clinical Research Center, the First Affiliate Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Ye Shen
- Department of Ophthalmology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| |
Collapse
|