1
|
Modrzejewska M, Zdanowska O. The Role of Heat Shock Protein 70 (HSP70) in the Pathogenesis of Ocular Diseases-Current Literature Review. J Clin Med 2024; 13:3851. [PMID: 38999417 PMCID: PMC11242833 DOI: 10.3390/jcm13133851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 06/23/2024] [Accepted: 06/28/2024] [Indexed: 07/14/2024] Open
Abstract
Heat shock proteins (HSPs) have been attracting the attention of researchers for many years. HSPs are a family of ubiquitous, well-characterised proteins that are generally regarded as protective multifunctional molecules that are expressed in response to different types of cell stress. Their activity in many organs has been reported, including the heart, brain, and retina. By acting as chaperone proteins, HSPs help to refold denatured proteins. Moreover, HSPs elicit inhibitory activity in apoptotic pathways and inflammation. Heat shock proteins were originally classified into several subfamilies, including the HSP70 family. The aim of this paper is to systematise information from the available literature about the presence of HSP70 in the human eye and its role in the pathogenesis of ocular diseases. HSP70 has been identified in the cornea, lens, and retina of a normal eye. The increased expression and synthesis of HSP70 induced by cell stress has also been demonstrated in eyes with pathologies such as glaucoma, eye cancers, cataracts, scarring of the cornea, ocular toxpoplasmosis, PEX, AMD, RPE, and diabetic retinopathy. Most of the studies cited in this paper confirm the protective role of HSP70. However, little is known about these molecules in the human eye and their role in the pathogenesis of eye diseases. Therefore, understanding the role of HSP70 in the pathophysiology of injuries to the cornea, lens, and retina is essential for the development of new therapies aimed at limiting and/or reversing the processes that cause damage to the eye.
Collapse
Affiliation(s)
- Monika Modrzejewska
- 2nd Department of Ophthalmology, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| | - Oliwia Zdanowska
- K. Marcinkowski University Hospital, 65-046 Zielona Góra, Poland
| |
Collapse
|
2
|
McAllister IL, Vijayasekaran S, McLenachan S, Bhikoo R, Chen FK, Zhang D, Kanagalingam E, Yu DY. Cytokine Levels in Experimental Branch Retinal Vein Occlusion Treated With Either Bevacizumab or Triamcinolone Acetonide. Transl Vis Sci Technol 2024; 13:13. [PMID: 38899953 PMCID: PMC11193067 DOI: 10.1167/tvst.13.6.13] [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: 01/24/2024] [Accepted: 05/01/2024] [Indexed: 06/21/2024] Open
Abstract
Purpose To compare gene expression changes following branch retinal vein occlusion (BRVO) in the pig with and without bevacizumab (BEV) and triamcinolone acetonide (TA). Methods Photothrombotic BRVOs were created in both eyes of four groups of nine pigs (2, 6, 10, and 20 days). In each group, six pigs received intravitreal injections of BEV in one eye and TA in the fellow eye, with three pigs serving as untreated BRVO controls. Three untreated pigs served as healthy controls. Expression of mRNA of vascular endothelial growth factor (VEGF), glial fibrillary acidic protein (GFAP), dystrophin (DMD), potassium inwardly rectifying channel subfamily J member 10 protein (Kir4.1, KCNJ10), aquaporin-4 (AQP4), stromal cell-derived factor-1α (CXCL12), interleukin-6 (IL6), interleukin-8 (IL8), monocyte chemoattractant protein-1 (CCL2), intercellular adhesion molecule 1 (ICAM1), and heat shock factor 1 (HSF1) were analyzed by quantitative reverse-transcription polymerase chain reaction. Retinal VEGF protein levels were characterized by immunohistochemistry. Results In untreated eyes, BRVO significantly increased expression of GFAP, IL8, CCL2, ICAM1, HSF1, and AQP4. Expression of VEGF, KCNJ10, and CXCL12 was significantly reduced by 6 days post-BRVO, with expression recovering to healthy control levels by day 20. Treatment with BEV or TA significantly increased VEGF, DMD, and IL6 expression compared with untreated BRVO eyes and suppressed BRVO-induced CCL2 and AQP4 upregulation, as well as recovery of KCNJ10 expression, at 10 to 20 days post-BRVO. Conclusions Inflammation and cellular osmohomeostasis rather than VEGF suppression appear to play important roles in BRVO-induced retinal neurodegeneration, enhanced in both BEV- and TA-treated retinas. Translational Relevance Inner retinal neurodegeneration seen in this acute model of BRVO appears to be mediated by inflammation and alterations in osmohomeostasis rather than VEGF inhibition, which may have implications for more specific treatment modalities in the acute phase of BRVO.
Collapse
Affiliation(s)
- Ian L. McAllister
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Western Australia, Australia
- Lions Eye Institute, Centre for Ophthalmology and Visual Science, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Sarojini Vijayasekaran
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Western Australia, Australia
- Lions Eye Institute, Centre for Ophthalmology and Visual Science, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Samuel McLenachan
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Western Australia, Australia
- Lions Eye Institute, Centre for Ophthalmology and Visual Science, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Riyaz Bhikoo
- Lions Eye Institute, Centre for Ophthalmology and Visual Science, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Fred K. Chen
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Western Australia, Australia
- Lions Eye Institute, Centre for Ophthalmology and Visual Science, The University of Western Australia, Nedlands, Western Australia, Australia
- Department of Ophthalmology, Royal Perth Hospital, Perth, Western Australia, Australia
- Ophthalmology, Department of Surgery, University of Melbourne, East Melbourne, Victoria, Australia
| | - Dan Zhang
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Western Australia, Australia
- Lions Eye Institute, Centre for Ophthalmology and Visual Science, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Emily Kanagalingam
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Western Australia, Australia
- Lions Eye Institute, Centre for Ophthalmology and Visual Science, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Dao-Yi Yu
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Western Australia, Australia
- Lions Eye Institute, Centre for Ophthalmology and Visual Science, The University of Western Australia, Nedlands, Western Australia, Australia
| |
Collapse
|
3
|
Yang J, Zhang X, Li Y, Yang N, Luo J, He T, Xing Y. Inhibition of TLR4/NF-κB pathway and endoplasmic reticulum stress by overexpressed S100A4 ameliorates retinal ischemia-reperfusion injury of mice. Mol Neurobiol 2024; 61:2228-2240. [PMID: 37872355 DOI: 10.1007/s12035-023-03709-w] [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: 07/22/2023] [Accepted: 10/11/2023] [Indexed: 10/25/2023]
Abstract
Retinal ischemia exists in various ischemic retinopathies including glaucoma, contributing to the death of retinal neurons. Calcium binding protein S100A4 is important in tumors, and our previous study found that S100A4 protects retinal ganglion cells (RGCs) against retinal ischemia-reperfusion (I/R) injury. This study was aimed to further discuss the neuroprotection and mechanisms of S100A4 in retinal I/R of mice. The rAAV-EF1α-s100a4-EGFP-WPRE or rAAV-EF1α-EGFP-WPRE-Pa was injected intravitreally 4 weeks before I/R. S100A4, molecules in TLR4 signaling pathway and endoplasmic reticulum (ER) stress branches, inflammatory molecules, and surviving RGCs and cholinergic amacrine (ChAT) cells were determined by quantitative PCR, western blot, or immunofluorescent staining. The apoptosis and necrosis of retinal neurons induced by I/R were inhibited by overexpressed S100A4. RGCs, ChAT cells, and the retinal function were preserved by S100A4 overexpressing 7 days after I/R. Mechanistically, the beneficial effects of S100A4 may be mediated by inhibiting the activation of TLR4 signaling pathway and alleviating ER stress, leading to the attenuation of inflammatory response of the retina after I/R. Our findings indicated that S100A4 has neuroprotective effect against retinal I/R injury, and promoting S100A4 expression may be an effective strategy to inhibit retinal neurons from degeneration in ischemic retinopathy.
Collapse
Affiliation(s)
- Jiayi Yang
- Ophthalmic Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiao Zhang
- Ophthalmic Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ying Li
- Ophthalmic Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ning Yang
- Ophthalmic Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jinyuan Luo
- Ophthalmic Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tao He
- Ophthalmic Center, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Yiqiao Xing
- Ophthalmic Center, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
4
|
Huang G, Zhang S, Liao J, Qin Y, Hong Y, Chen Q, Lin Y, Li Y, Lan L, Hu W, Huang K, Tang F, Tang N, Jiang L, Shen C, Cui L, Zhong H, Li M, Lu P, Shu Q, Wei Y, Xu F. BMX deletion mitigates neuroinflammation induced by retinal ischemia/reperfusion through modulation of the AKT/ERK/STAT3 signaling cascade. Heliyon 2024; 10:e27114. [PMID: 38434304 PMCID: PMC10907772 DOI: 10.1016/j.heliyon.2024.e27114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/13/2024] [Accepted: 02/23/2024] [Indexed: 03/05/2024] Open
Abstract
Aims Retinal ischemia/reperfusion (I/R) injury is implicated in the etiology of various ocular disorders. Prior research has demonstrated that bone marrow tyrosine kinase on chromosome X (BMX) contributes to the advancement of ischemic disease and inflammatory reactions. Consequently, the current investigation aims to evaluate BMX's impact on retinal I/R injury and clarify its implied mechanism of action. Main methods This study utilized male and female systemic BMX knockout (BMX-/-) mice to conduct experiments. The utilization of Western blot assay and immunofluorescence labeling techniques was employed to investigate variations in the expression of protein and tissue localization. Histomorphological changes were observed through H&E staining and SD-OCT examination. Visual function changes were assessed through electrophysiological experiments. Furthermore, apoptosis in the retina was identified using the TUNEL assay, as well as the ELISA technique, which has been utilized to determine the inflammatory factors level. Key findings Our investigation results revealed that the knockdown of BMX did not yield a significant effect on mouse retina. In mice, BMX knockdown mitigated the negative impact of I/R injury on retinal tissue structure and visual function. BMX knockdown effectively reduced apoptosis, suppressed inflammatory responses, and decreased inflammatory factors subsequent to I/R injury. The outcomes of the current investigation revealed that BMX knockdown partially protected the retina through downregulating phosphorylation of AKT/ERK/STAT3 pathway. Significance Our investigation showed that BMX-/- reduces AKT, ERK, and STAT3 phosphorylation, reducing apoptosis and inflammation. Thus, this strategy protected the retina from structural and functional damage after I/R injury.
Collapse
Affiliation(s)
- Guangyi Huang
- Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology &Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, 530021, Guangxi, China
| | - Shaoyang Zhang
- Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology &Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, 530021, Guangxi, China
| | - Jing Liao
- Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology &Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, 530021, Guangxi, China
| | - Yuanjun Qin
- Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology &Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, 530021, Guangxi, China
| | - Yiyi Hong
- Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology &Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, 530021, Guangxi, China
| | - Qi Chen
- Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology &Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, 530021, Guangxi, China
| | - Yunru Lin
- Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology &Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, 530021, Guangxi, China
| | - Yue Li
- Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology &Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, 530021, Guangxi, China
| | - Lin Lan
- Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology &Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, 530021, Guangxi, China
| | - Wen Hu
- Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology &Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, 530021, Guangxi, China
| | - Kongqian Huang
- Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology &Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, 530021, Guangxi, China
| | - Fen Tang
- Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology &Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, 530021, Guangxi, China
| | - Ningning Tang
- Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology &Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, 530021, Guangxi, China
| | - Li Jiang
- Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology &Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, 530021, Guangxi, China
| | - Chaolan Shen
- Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology &Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, 530021, Guangxi, China
| | - Ling Cui
- Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology &Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, 530021, Guangxi, China
| | - Haibin Zhong
- Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology &Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, 530021, Guangxi, China
| | - Min Li
- Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology &Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, 530021, Guangxi, China
| | - Peng Lu
- Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology &Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, 530021, Guangxi, China
| | - Qinmeng Shu
- Eye Institute, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Key Laboratory of Myopia (Fudan University), Chinese Academy of Medical Sciences, National Health Commission, Shanghai, China
| | - Yantao Wei
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 7 Jinsui Road, Guangzhou, 510060, China
| | - Fan Xu
- Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology &Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, 530021, Guangxi, China
| |
Collapse
|
5
|
Hu C, Feng Y, Huang G, Cui K, Fan M, Xiang W, Shi Y, Ye D, Ye H, Bai X, Xu F, Xu Y, Huang J. Melatonin prevents EAAC1 deletion-induced retinal ganglion cell degeneration by inhibiting apoptosis and senescence. J Pineal Res 2024; 76:e12916. [PMID: 37786968 DOI: 10.1111/jpi.12916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 08/28/2023] [Accepted: 09/14/2023] [Indexed: 10/04/2023]
Abstract
Normal tension glaucoma (NTG) is referred to as a progressive degenerative disorder of the retinal ganglion cells (RGCs), resulting in nonreversible visual defects, despite intraocular pressure levels within the statistically normal range. Current therapeutic strategies for NTG yield limited benefits. Excitatory amino acid carrier 1 (EAAC1) knockout (EAAC1-/- ) in mice has been shown to induce RGC degeneration without elevating intraocular pressure, mimicking pathological characteristics of NTG. In this study, we explored whether daily oral administration of melatonin could block RGCs loss and prevent retinal morphology and function defects associated with EAAC1 deletion. We also explored the molecular mechanisms underlying EAAC1 deletion-induced RGC degeneration and the neuroprotective effects of melatonin. Our RNA sequencing and in vivo data indicated EAAC1 deletion caused elevated oxidative stress, activation of apoptosis and cellular senescence pathways, and neuroinflammation in RGCs. However, melatonin administration efficiently prevented these detrimental effects. Furthermore, we investigated the potential role of apoptosis- and senescence-related redox-sensitive factors in EAAC1 deletion-induced RGCs degeneration and the neuroprotective effects of melatonin administration. We observed remarkable upregulation of p53, whereas NRF2 and Sirt1 expression were significantly decreased in EAAC1-/- mice, which were prevented by melatonin treatment, suggesting that melatonin exerted its neuroprotective effects possibly through modulating NRF2/p53/Sirt1 redox-sensitive signaling pathways. Overall, our study provided a solid foundation for the application of melatonin in the management of NTG.
Collapse
Affiliation(s)
- Chenyang Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yanlin Feng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Guangyi Huang
- Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology, Department of Ophthalmology, The People's Hospital of Guangxi Zhuang Autonomous Region, Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, China
| | - Kaixuan Cui
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Matthew Fan
- Yale College, Yale University, New Haven, Connecticut, USA
| | - Wu Xiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yuxun Shi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Dan Ye
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Huiwen Ye
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xue Bai
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Fan Xu
- Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology, Department of Ophthalmology, The People's Hospital of Guangxi Zhuang Autonomous Region, Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, China
| | - Yue Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jingjing Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| |
Collapse
|
6
|
Buscho SE, Xia F, Shi S, Lin JL, Szczesny B, Zhang W, Motamedi M, Liu H. Non-Invasive Evaluation of Retinal Vascular Alterations in a Mouse Model of Optic Neuritis Using Laser Speckle Flowgraphy and Optical Coherence Tomography Angiography. Cells 2023; 12:2685. [PMID: 38067113 PMCID: PMC10705764 DOI: 10.3390/cells12232685] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/04/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
Optic neuritis, a characteristic feature of multiple sclerosis (MS), involves the inflammation of the optic nerve and the degeneration of retinal ganglion cells (RGCs). Although previous studies suggest that retinal blood flow alterations occur during optic neuritis, the precise location, the degree of impairment, and the underlying mechanisms remain unclear. In this study, we utilized two emerging non-invasive imaging techniques, laser speckle flowgraphy (LSFG) and optical coherence tomography angiography (OCTA), to investigate retinal vascular changes in a mouse model of MS, known as experimental autoimmune encephalomyelitis (EAE). We associated these changes with leukostasis, RGC injury, and the overall progression of EAE. LSFG imaging revealed a progressive reduction in retinal blood flow velocity and increased vascular resistance near the optic nerve head in the EAE model, indicating impaired ocular blood flow. OCTA imaging demonstrated significant decreases in vessel density, number of junctions, and total vessel length in the intermediate and deep capillary plexus of the EAE mice. Furthermore, our analysis of leukostasis revealed a significant increase in adherent leukocytes in the retinal vasculature of the EAE mice, suggesting the occurrence of vascular inflammation in the early development of EAE pathology. The abovechanges preceded or were accompanied by the characteristic hallmarks of optic neuritis, such as RGC loss and reduced visual acuity. Overall, our study sheds light on the intricate relationship between retinal vascular alterations and the progression of optic neuritis as well as MS clinical score. It also highlights the potential for the development of image-based biomarkers for the diagnosis and monitoring of optic neuritis as well as MS, particularly in response to emerging treatments.
Collapse
Affiliation(s)
- Seth E. Buscho
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA; (S.E.B.); (F.X.); (S.S.); (J.L.L.); (B.S.); (W.Z.); (M.M.)
| | - Fan Xia
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA; (S.E.B.); (F.X.); (S.S.); (J.L.L.); (B.S.); (W.Z.); (M.M.)
| | - Shuizhen Shi
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA; (S.E.B.); (F.X.); (S.S.); (J.L.L.); (B.S.); (W.Z.); (M.M.)
| | - Jonathan L. Lin
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA; (S.E.B.); (F.X.); (S.S.); (J.L.L.); (B.S.); (W.Z.); (M.M.)
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Bartosz Szczesny
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA; (S.E.B.); (F.X.); (S.S.); (J.L.L.); (B.S.); (W.Z.); (M.M.)
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Wenbo Zhang
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA; (S.E.B.); (F.X.); (S.S.); (J.L.L.); (B.S.); (W.Z.); (M.M.)
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Massoud Motamedi
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA; (S.E.B.); (F.X.); (S.S.); (J.L.L.); (B.S.); (W.Z.); (M.M.)
| | - Hua Liu
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA; (S.E.B.); (F.X.); (S.S.); (J.L.L.); (B.S.); (W.Z.); (M.M.)
| |
Collapse
|
7
|
Zhao WJ, Fan CL, Hu XM, Ban XX, Wan H, He Y, Zhang Q, Xiong K. Regulated Cell Death of Retinal Ganglion Cells in Glaucoma: Molecular Insights and Therapeutic Potentials. Cell Mol Neurobiol 2023; 43:3161-3178. [PMID: 37338781 DOI: 10.1007/s10571-023-01373-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 06/05/2023] [Indexed: 06/21/2023]
Abstract
Glaucoma is a group of diseases characterized by the degeneration of retinal ganglion cells (RGCs) and progressive, irreversible vision loss. High intraocular pressure (IOP) heightens the likelihood of glaucoma and correlates with RGC loss. While the current glaucoma therapy prioritizes lower the IOP; however, RGC, and visual loss may persist even when the IOP is well-controlled. As such, discovering and creating IOP-independent neuroprotective strategies for safeguard RGCs is crucial for glaucoma management. Investigating and clarifying the mechanism behind RGC death to counteract its effects is a promising direction for glaucoma control. Empirical studies of glaucoma reveal the role of multiple regulated cell death (RCD) pathways in RGC death. This review delineates the RCD of RGCs following IOP elevation and optic nerve damage and discusses the substantial benefits of mitigating RCD in RGCs in preserving visual function.
Collapse
Affiliation(s)
- Wen-Juan Zhao
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Hunan Province, No. 172, Tongzipo Road, Yuelu District, Changsha City, 410013, China
| | - Chun-Ling Fan
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Hunan Province, No. 172, Tongzipo Road, Yuelu District, Changsha City, 410013, China
| | - Xi-Min Hu
- Department of Dermatology, Xiangya Hospital, Central South University, Hunan Province, No. 172, Tongzipo Road, Yuelu District, Changsha City, 410013, China
| | - Xiao-Xia Ban
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Hunan Province, No. 172, Tongzipo Road, Yuelu District, Changsha City, 410013, China
| | - Hao Wan
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Hunan Province, No. 172, Tongzipo Road, Yuelu District, Changsha City, 410013, China
| | - Ye He
- Changsha Aier Eye Hospital, Hunan Province, No. 188, Furong Road, Furong District, Changsha City, 410015, China
| | - Qi Zhang
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Hunan Province, No. 172, Tongzipo Road, Yuelu District, Changsha City, 410013, China.
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, 571199, China.
| | - Kun Xiong
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Hunan Province, No. 172, Tongzipo Road, Yuelu District, Changsha City, 410013, China.
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, 571199, China.
- Hunan Key Laboratory of Ophthalmology, Changsha, 410013, China.
| |
Collapse
|
8
|
Shi S, Ding C, Zhu S, Xia F, Buscho SE, Li S, Motamedi M, Liu H, Zhang W. PERK Inhibition Suppresses Neovascularization and Protects Neurons During Ischemia-Induced Retinopathy. Invest Ophthalmol Vis Sci 2023; 64:17. [PMID: 37566408 PMCID: PMC10424802 DOI: 10.1167/iovs.64.11.17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 07/10/2023] [Indexed: 08/12/2023] Open
Abstract
Purpose Retinal ischemia is a common cause of a variety of eye diseases, such as retinopathy of prematurity, diabetic retinopathy, and vein occlusion. Protein kinase RNA-activated-like endoplasmic reticulum (ER) kinase (PERK), one of the main ER stress sensor proteins, has been involved in many diseases. In this study, we investigated the role of PERK in ischemia-induced retinopathy using a mouse model of oxygen-induced retinopathy (OIR). Methods OIR was induced by subjecting neonatal pups to 70% oxygen at postnatal day 7 (P7) followed by returning to room air at P12. GSK2606414, a selective PERK inhibitor, was orally administrated to pups right after they were returned to room air once daily until 1 day before sample collection. Western blot, immunostaining, and quantitative PCR were used to assess PERK phosphorylation, retinal changes, and signaling pathways in relation to PERK inhibition. Results PERK phosphorylation was prominently increased in OIR retinas, which was inhibited by GSK2606414. Concomitantly, PERK inhibition significantly reduced retinal neovascularization (NV) and retinal ganglion cell (RGC) loss, restored astrocyte network, and promoted revascularization. Furthermore, PERK inhibition downregulated the recruitment/proliferation of mononuclear phagocytes but did not affect OIR-upregulated canonical angiogenic pathways. Conclusions Our results demonstrate that PERK is involved in ischemia-induced retinopathy and its inhibition using GSK2606414 could offer an effective therapeutic intervention aimed at alleviating retinal NV while preventing neuron loss during retinal ischemia.
Collapse
Affiliation(s)
- Shuizhen Shi
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
| | - Chun Ding
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
| | - Shuang Zhu
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
| | - Fan Xia
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
| | - Seth E. Buscho
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
| | - Shengguo Li
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
| | - Massoud Motamedi
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
| | - Hua Liu
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
| | - Wenbo Zhang
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
- Departments of Neurobiology, University of Texas Medical Branch, Galveston, Texas, United States
| |
Collapse
|
9
|
Hu XM, Li ZX, Deng J, Han Y, Lu S, Zhang Q, Luo ZQ, Xiong K. Integration of Theory and Practice in Medical Morphology Curriculum in Postgraduate Training: A Flipped Classroom and Case-based Learning Exercise. Curr Med Sci 2023; 43:741-748. [PMID: 37455278 DOI: 10.1007/s11596-023-2759-9] [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: 01/20/2023] [Accepted: 02/27/2023] [Indexed: 07/18/2023]
Abstract
OBJECTIVE The integration of training in theory and practice across the medical education spectrum is being encouraged to increase student understanding and skills in the sciences. This study aimed to determine the deciding factors that drive students' perceived advantages in class to improve precision education and the teaching model. METHODS A mixed strategy of an existing flipped classroom (FC) and a case-based learning (CBL) model was conducted in a medical morphology curriculum for 575 postgraduate students. The subjective learning evaluation of the individuals (learning time, engagement, study interest and concentration, and professional integration) was collected and analyzed after FC-CBL model learning. RESULTS The results from the general evaluation showed promising results of the medical morphology in the FC-CBL model. Students felt more engaged by instructors in person and benefited in terms of time-saving, flexible arrangements, and professional improvement. Our study contributed to the FC-CBL model in Research Design in postgraduate training in 4 categories: 1) advancing a guideline of precision teaching according to individual characteristics; 2) revealing whether a learning background is needed for a Research Design course to guide setting up a preliminary course; 3) understanding the perceived advantages and their interfaces; and 4) barriers and/or improvement to implement the FC-CBL model in the Research Design class, such as a richer description of e-learning and hands-on practice. CONCLUSION Undertaking a FC-CBL combined model could be a useful addition to pedagogy for medical morphology learning in postgraduate training.
Collapse
Affiliation(s)
- Xi-Min Hu
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410000, China
| | - Zhi-Xin Li
- Clinical Medicine Eight-year Program, 02 Class, 18 Grade, Xiangya School of Medicine, Central South University, Changsha, 410000, China
| | - Jing Deng
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, 410000, China
| | - Yang Han
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, 410008, China
| | - Shuang Lu
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410000, China
| | - Qi Zhang
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410000, China
| | - Zi-Qiang Luo
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, 410008, China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410000, China.
- Hunan Key Laboratory of Ophthalmology, Changsha, 410000, China.
- Key Laboratory of Emergency and Trauma, Ministry of Education, College of Emergency and Trauma, Hainan Medical University, Haikou, 016000, China.
| |
Collapse
|
10
|
He Q, Xiao L, Shi Y, Li W, Xin X. Natural products: protective effects against ischemia-induced retinal injury. Front Pharmacol 2023; 14:1149708. [PMID: 37180697 PMCID: PMC10169696 DOI: 10.3389/fphar.2023.1149708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 04/05/2023] [Indexed: 05/16/2023] Open
Abstract
Ischemic retinal damage, a common condition associated with retinal vascular occlusion, glaucoma, diabetic retinopathy, and other eye diseases, threatens the vision of millions of people worldwide. It triggers excessive inflammation, oxidative stress, apoptosis, and vascular dysfunction, leading to the loss and death of retinal ganglion cells. Unfortunately, minority drugs are available for treating retinal ischemic injury diseases, and their safety are limited. Therefore, there is an urgent need to develop more effective treatments for ischemic retinal damage. Natural compounds have been reported to have antioxidant, anti-inflammatory, and antiapoptotic properties that can be used to treat ischemic retinal damage. In addition, many natural compounds have been shown to exhibit biological functions and pharmacological properties relevant to the treatment of cellular and tissue damage. This article reviews the neuroprotective mechanisms of natural compounds involve treating ischemic retinal injury. These natural compounds may serve as treatments for ischemia-induced retinal diseases.
Collapse
Affiliation(s)
- Qianxiong He
- Department of Ophthalmology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Liuyi Xiao
- Department of Ophthalmology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yuanjiang Shi
- Department of Ophthalmology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Clinical Medicine School of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan, China
| | - Wanrong Li
- Department of Ophthalmology, People's Hospital of Golog Tibetan Autonomous Prefecture, Golog, Qinghai, China
| | - Xiaorong Xin
- Department of Ophthalmology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
11
|
Belenichev IF, Aliyeva OG, Popazova OO, Bukhtiyarova NV. Involvement of heat shock proteins HSP70 in the mechanisms of endogenous neuroprotection: the prospect of using HSP70 modulators. Front Cell Neurosci 2023; 17:1131683. [PMID: 37138769 PMCID: PMC10150069 DOI: 10.3389/fncel.2023.1131683] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 03/28/2023] [Indexed: 05/05/2023] Open
Abstract
This analytical review summarizes literature data and our own research on HSP70-dependent mechanisms of neuroprotection and discusses potential pharmacological agents that can influence HSP70 expression to improve neurological outcomes and effective therapy. The authors formed a systemic concepts of the role of HSP70-dependent mechanisms of endogenous neuroprotection aimed at stopping the formation of mitochondrial dysfunction, activation of apoptosis, desensitization of estrogen receptors, reduction of oxidative and nitrosative stress, prevention of morpho-functional changes in brain cells during cerebral ischemia, and experimentally substantiated new target links for neuroprotection. Heat shock proteins (HSPs) are an evolutionarily integral part of the functioning of all cells acting as intracellular chaperones that support cell proteostasis under normal and various stress conditions (hyperthermia, hypoxia, oxidative stress, radiation, etc.). The greatest curiosity in conditions of ischemic brain damage is the HSP70 protein, as an important component of the endogenous neuroprotection system, which, first of all, performs the function of intracellular chaperones and ensures the processes of folding, holding and transport of synthesized proteins, as well as their degradation, both under normoxic conditions and stress-induced denaturation. A direct neuroprotective effect of HSP70 has been established, which is realized through the regulation the processes of apoptosis and cell necrosis due to a long-term effect on the synthesis of antioxidant enzymes, chaperone activity, and stabilization of active enzymes. An increase in the level of HSP70 leads to the normalization of the glutathione link of the thiol-disulfide system and an increase in the resistance of cells to ischemia. HSP 70 is able to activate and regulate compensatory ATP synthesis pathways during ischemia. It was found that in response to the cerebral ischemia formation, HIF-1a is expressed, which initiates the launch of compensatory mechanisms for energy production. Subsequently, the regulation of these processes switches to HSP70, which "prolongs" the action of HIF-1a, and also independently maintains the expression of mitochondrial NAD-dependent malate dehydrogenase activity, thereby maintaining the activity of the malate-aspartate shuttle mechanism for a long time. During ischemia of organs and tissues, HSP70 performs a protective function, which is realized through increased synthesis of antioxidant enzymes, stabilization of oxidatively damaged macromolecules, and direct anti-apoptotic and mitoprotective action. Such a role of these proteins in cellular reactions during ischemia raises the question of the development of new neuroprotective agents which are able to provide modulation/protection of the genes encoding the synthesis of HSP 70 and HIF-1a proteins. Numerous studies of recent years have noted the important role of HSP70 in the implementation of the mechanisms of metabolic adaptation, neuroplasticity and neuroprotection of brain cells, so the positive modulation of the HSP70 system is a perspective concept of neuroprotection, which can improve the efficiency of the treatment of ischemic-hypoxic brain damage and be the basis for substantiating of the feasibility of using of HSP70 modulators as promising neuroprotectors.
Collapse
Affiliation(s)
- Igor F. Belenichev
- Department of Pharmacology and Medical Formulation With Course of Normal Physiology, Zaporizhzhia State Medical University, Zaporizhzhia, Ukraine
| | - Olena G. Aliyeva
- Department of Medical Biology, Parasitology and Genetics, Zaporizhzhia State Medical University, Zaporizhzhia, Ukraine
| | - Olena O. Popazova
- Department of Histology, Cytology and Embryology, Zaporizhzhia State Medical University, Zaporizhzhia, Ukraine
| | - Nina V. Bukhtiyarova
- Department of Clinical Laboratory Diagnostics, Zaporizhzhia State Medical University, Zaporizhzhia, Ukraine
| |
Collapse
|
12
|
Specific Activation of Yamanaka Factors via HSF1 Signaling in the Early Stage of Zebrafish Optic Nerve Regeneration. Int J Mol Sci 2023; 24:ijms24043253. [PMID: 36834675 PMCID: PMC9961437 DOI: 10.3390/ijms24043253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/30/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
In contrast to the case in mammals, the fish optic nerve can spontaneously regenerate and visual function can be fully restored 3-4 months after optic nerve injury (ONI). However, the regenerative mechanism behind this has remained unknown. This long process is reminiscent of the normal development of the visual system from immature neural cells to mature neurons. Here, we focused on the expression of three Yamanaka factors (Oct4, Sox2, and Klf4: OSK), which are well-known inducers of induced pluripotent stem (iPS) cells in the zebrafish retina after ONI. mRNA expression of OSK was rapidly induced in the retinal ganglion cells (RGCs) 1-3 h after ONI. Heat shock factor 1 (HSF1) mRNA was most rapidly induced in the RGCs at 0.5 h. The activation of OSK mRNA was completely suppressed by the intraocular injection of HSF1 morpholino prior to ONI. Furthermore, the chromatin immunoprecipitation assay showed the enrichment of OSK genomic DNA bound to HSF1. The present study clearly showed that the rapid activation of Yamanaka factors in the zebrafish retina was regulated by HSF1, and this sequential activation of HSF1 and OSK might provide a key to unlocking the regenerative mechanism of injured RGCs in fish.
Collapse
|
13
|
Mei T, Wu J, Wu K, Zhao M, Luo J, Liu X, Shang B, Xu W, Yang Z, Lai Y, Liu C, Gong H, Gao X, Zhuo Y, Lin M, Zhao L. Lipocalin 2 induces visual impairment by promoting ferroptosis in retinal ischemia-reperfusion injury. ANNALS OF TRANSLATIONAL MEDICINE 2023; 11:3. [PMID: 36760251 PMCID: PMC9906199 DOI: 10.21037/atm-22-3298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 11/07/2022] [Indexed: 01/07/2023]
Abstract
Background Retinal ischemia-reperfusion (RIR) is a common pathological condition that can lead to retinal ganglion cell (RGC) death and visual impairment. However, the pathogenesis of RGC loss and visual impairment caused by retinal ischemia remains unclear. Methods A mouse model of elevated intraocular pressure (IOP)-induced RIR injury was used. Flash visual evoked potentials (FVEPs) and electroretinography (ERG) recordings were performed to assess visual function. The structural integrity of the retina and the number of RGC were assessed using hematoxylin and eosin (HE) staining and retinal flat mounts. Ferroptosis was evaluated by testing the levels of glutathione (GSH), malondialdehyde (MDA), glutathione peroxidase (GPX4), and ferritin light chains (FTL) in the retina of wild-type (WT) and lipocalin-2 transgenic (LCN2-TG) mice after RIR injury. Results We found that LCN2 was mainly expressed in the RGC layer in the retina of wild-type mice and remarkably upregulated after RIR injury. Compared with wild-type mice, aggravated RGC death and visual impairment were exhibited in LCN2-TG mice with RIR injury. Moreover, LCN2 overexpression activated glial cells and upregulated proinflammatory factors. More importantly, we found that LCN2 strongly promoted ferroptosis signaling in RGC death and visual impairment. Liproxstatin-1, an inhibitor of ferroptosis, could significantly ameliorate RGC death and visual impairment. Furthermore, we found significantly alleviated RGC death and retinal damage in LCN2 heterozygous knockout mice. Conclusions Our study provides important insights linking upregulated LCN2-mediated promotion of ferroptosis to RGC death and visual function impairment in the pathogenesis of ischemic retinopathy.
Collapse
Affiliation(s)
- Tingfang Mei
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jinwen Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Keling Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Minglei Zhao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jingyi Luo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xinqi Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China;,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Bizhi Shang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Wenchang Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Zeqiu Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yuhua Lai
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Chujun Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Haijun Gong
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China;,Department of Ophthalmology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xinbo Gao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yehong Zhuo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Mingkai Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Ling Zhao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| |
Collapse
|
14
|
Abstract
Purpose: Retinal ganglion cell death occurs during the glaucoma pathological process, and it is significant because of the poor regeneration capacity of retinal ganglion cells. With a constantly increasing understanding of retinal cell death mechanisms, we now know that simply blocking a specific mechanism of cell death might not prevent retinal ganglion cell death. This review aimed to summarize the mechanisms of retinal cell death in glaucoma models and discuss the caveats in restoring visual function in these studies.Methods: A literature search was done on PubMed using key words including glaucoma, ocular hypertension, retinal ganglion cell, cell death, apoptosis, necroptosis, pyroptosis, ferroptosis, autophagic cell death, and parthanatos. The literature was reviewed to summarize the information about the lethal pathways of retinal ganglion cell in the glaucoma-like animal models.Results: Based on the purpose, 100 studies were selected and discussed in this review.Conclusions: The damage to ganglion cells in glaucoma-like animals can occur via multiple lethal pathways and the molecular mechanisms are still incompletely understood. Further investigations on the crosstalk between different cell death pathways and the common upstream regulators could augment the development of novel targeting agents for the curative treatment of glaucoma.
Collapse
Affiliation(s)
- Yuting Yang
- Department of Ophthalmology & Visual Science, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xinghuai Sun
- Department of Ophthalmology & Visual Science, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,NHC Key Laboratory of Myopia, Chinese Academy of Medical Sciences, and Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China.,State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Zhang Q, Xiong K, Yan WT, Zhao WJ, Hu XM, Ban XX, Ning WY, Wan H. PANoptosis-like cell death in ischemia/reperfusion injury of retinal neurons. Neural Regen Res 2023; 18:357-363. [PMID: 35900430 PMCID: PMC9396479 DOI: 10.4103/1673-5374.346545] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
16
|
Zhu MX, Ma XF, Niu X, Fan GB, Li Y. Mitochondrial unfolded protein response in ischemia-reperfusion injury. Brain Res 2022; 1797:148116. [PMID: 36209898 DOI: 10.1016/j.brainres.2022.148116] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/27/2022] [Accepted: 10/02/2022] [Indexed: 11/21/2022]
Abstract
Mitochondrial unfolded protein response (UPRmt) is a mitochondrial stress response that activates the transcriptional program of mitochondrial chaperone proteins and proteases to keep protein homeostasis in mitochondria. Ischemia-reperfusion injury results in multiple severe clinical issues linked to high morbidity and mortality in various disorders. The pathophysiology and pathogenesis of ischemia-reperfusion injury are complex and multifactorial. Emerging evidence showed the roles of UPRmt signaling in ischemia-reperfusion injury. Herein, we discuss the regulatory mechanisms underlying UPRmt signaling in C. elegans and mammals. Furthermore, we review the recent studies into the roles and mechanisms of UPRmt signaling in ischemia-reperfusion injury of the heart, brain, kidney, and liver. Further research of UPRmt signaling will potentially develop novel therapeutic strategies against ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Ming-Xi Zhu
- Department of Anatomy, School of Basic Medicine and Life Science, Hainan Medical University, Hainan, China
| | - Xiao-Fei Ma
- Department of ICU, The 4th Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xing Niu
- Department of Second Clinical College, Shengjing Hospital of China Medical University, Shenyang, China
| | - Gui-Bo Fan
- Department of Anesthesiology, The 4th Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Yan Li
- Department of Anesthesiology, The 4th Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
17
|
Ye D, Xu Y, Shi Y, Fan M, Lu P, Bai X, Feng Y, Hu C, Cui K, Tang X, Liao J, Huang W, Xu F, Liang X, Huang J. Anti-PANoptosis is involved in neuroprotective effects of melatonin in acute ocular hypertension model. J Pineal Res 2022; 73:e12828. [PMID: 36031799 DOI: 10.1111/jpi.12828] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/12/2022] [Accepted: 08/24/2022] [Indexed: 11/29/2022]
Abstract
Acute ocular hypertension (AOH) is the most important characteristic of acute glaucoma, which can lead to retinal ganglion cell (RGC) death and permanent vision loss. So far, approved effective therapy is still lacking in acute glaucoma. PANoptosis (pyroptosis, apoptosis, and necroptosis), which consists of three key modes of programmed cell death-apoptosis, necroptosis, and pyroptosis-may contribute to AOH-induced RGC death. Previous studies have demonstrated that melatonin (N-acetyl-5-methoxytryptamine) exerts a neuroprotective effect in many retinal degenerative diseases. However, whether melatonin is anti-PANoptotic and neuroprotective in the progression of acute glaucoma remains unclear. Thus, this study aimed to explore the role of melatonin in AOH retinas and its underlying mechanisms. The results showed that melatonin treatment attenuated the loss of ganglion cell complex thickness, retinal nerve fiber layer thickness, and RGC after AOH injury, and improved the amplitudes of a-wave, b-wave, and oscillatory potentials in the electroretinogram. Additionally, the number of terminal deoxynucleotidyl transferase dUTP nick-end labeling-positive cells was decreased, and the upregulation of cleaved caspase-8, cleaved caspase-3, Bax, and Bad and downregulation of Bcl-2 and p-Bad were inhibited after melatonin administration. Meanwhile, both the expression and activation of MLKL, RIP1, and RIP3, along with the number of PI-positive cells, were reduced in melatonin-treated mice, and p-RIP3 was in both RGC and microglia/macrophage after AOH injury. Furthermore, melatonin reduced the expression of NLRP3, ASC, cleaved caspase-1, gasdermin D (GSDMD), and cleaved GSDMD, and decreased the number of Iba1/interleukin-1β-positive cells. In conclusion, melatonin ameliorated retinal structure, prevented retinal dysfunction after AOH, and exerted a neuroprotective effect via inhibition of PANoptosis in AOH retinas.
Collapse
Affiliation(s)
- Dan Ye
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yue Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yuxun Shi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Matthew Fan
- Yale College, Yale University, New Haven, Connecticut, USA
| | - Peng Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xue Bai
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yanlin Feng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Chenyang Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Kaixuan Cui
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xiaoyu Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jing Liao
- Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences & Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology, Nanning, China
| | - Wei Huang
- Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences & Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology, Nanning, China
| | - Fan Xu
- Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences & Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology, Nanning, China
| | - Xiaoling Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jingjing Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| |
Collapse
|
18
|
Wang S, Yu A, Han M, Chen X, Li Z, Ke M, Cai X, Ai M, Xing Y. Pathological Changes and Expression of JAK-STAT Signaling Pathway Hallmark Proteins in Rat Retinas at Different Time Points After Retinal Ischemia Reperfusion Injury. Pathol Oncol Res 2022; 28:1610385. [PMID: 35515015 PMCID: PMC9061953 DOI: 10.3389/pore.2022.1610385] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 03/30/2022] [Indexed: 11/25/2022]
Abstract
Retinal ischemia reperfusion injury (RIRI) is a conventional pathological process in various retinal vascular diseases. Many studies select only one specific time point to apply drugs and then assess the therapeutic effect of drugs; however, the baselines are not the same at different time points, which may cause variation in the judgement. Therefore, further investigation is needed. Accordingly, this study aimed to investigate the pathological changes of retinal structure, expression of JAK-STAT signaling pathway hallmark proteins, and apoptosis at different time points after retinal ischemia reperfusion injury in rats. Sixty-six male SPF Sprague-Dawley rats were randomly divided into six groups: control group, RIRI 0, 6-, 24-, 72-, and 144-h groups. RIRI models were induced by perfusing equilibrium solution into the right eye anterior chamber to increase intraocular pressure to 110 mmHg for 60 min. Rats were sacrificed at different time points after reperfusion. Then hematoxylin-eosin staining, transmission electron microscope, immunohistochemistry, western blot, and TUNEL were used. Hematoxylin-eosin showed the pathological changes while transmission electron microscope revealed the ultra-structure changes of retina after RIRI. Immunohistochemistry showed that JAK2, STAT3, p-JAK2, p-STAT3, Bax, and Bcl-2 proteins mainly located in ganglion cell layer and inner nuclear layer, the relative expression of former five proteins had significant differences vs. control group (p < 0.05), while Bcl-2 had no significant difference. In western blot, the protein expressing of JAK2, STAT3, p-JAK2, p-STAT3, p-Akt, and Bax had significant differences vs. control group (p < 0.05), while Akt and Bcl-2 had no significant differences. TUNEL staining showed the number of apoptosis positive cells rose initially but declined later, with a peak value at RIRI 24 h group. The dynamic changes of hallmark proteins at different time points after RIRI indicate that JAK-STAT signaling pathway activates rapidly but weakens later and plays a vital role in RIRI, and apoptosis is involved in RIRI with a peak value at 24 h in the process, suggesting a potential therapeutic direction and time window for treating RIRI.
Collapse
Affiliation(s)
- Shun Wang
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Aihua Yu
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Mengyao Han
- Retinal and Vitreous Diseases Department, Wuhan Aier Eye Hospital of Wuhan University, Wuhan, China
| | - Xiaomin Chen
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhi Li
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Min Ke
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiaojun Cai
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ming Ai
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yiqiao Xing
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
19
|
Role of Curcumin in Retinal Diseases-A review. Graefes Arch Clin Exp Ophthalmol 2022; 260:1457-1473. [PMID: 35015114 PMCID: PMC8748528 DOI: 10.1007/s00417-021-05542-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 12/21/2021] [Accepted: 12/27/2021] [Indexed: 02/08/2023] Open
Abstract
PURPOSE To review the role of curcumin in retinal diseases, COVID era, modification of the molecule to improve bioavailability and its future scope. METHODS PubMed and MEDLINE searches were pertaining to curcumin, properties of curcumin, curcumin in retinal diseases, curcumin in diabetic retinopathy, curcumin in age-related macular degeneration, curcumin in retinal and choroidal diseases, curcumin in retinitis pigmentosa, curcumin in retinal ischemia reperfusion injury, curcumin in proliferative vitreoretinopathy and curcumin in current COVID era. RESULTS In experimental models, curcumin showed its pleiotropic effects in retinal diseases like diabetic retinopathy by increasing anti-oxidant enzymes, upregulating HO-1, nrf2 and reducing or inhibiting inflammatory mediators, growth factors and by inhibiting proliferation and migration of retinal endothelial cells in a dose-dependent manner in HRPC, HREC and ARPE-19 cells. In age-related macular degeneration, curcumin acts by reducing ROS and inhibiting apoptosis inducing proteins and cellular inflammatory genes and upregulating HO-1, thioredoxin and NQO1. In retinitis pigmentosa, curcumin has been shown to delay structural defects of P23H gene in P23H-rhodopsin transgenic rats. In proliferative vitreoretinopathy, curcumin inhibited the action of EGF in a dose- and time-dependent manner. In retinal ischemia reperfusion injury, curcumin downregulates IL-17, IL-23, NFKB, STAT-3, MCP-1 and JNK. In retinoblastoma, curcumin inhibits proliferation, migration and apoptosis of RBY79 and SO-RB50. Curcumin has already proven its efficacy in inhibiting viral replication, coagulation and cytokine storm in COVID era. CONCLUSION Curcumin is an easily available spice used traditionally in Indian cooking. The benefits of curcumin are manifold, and large randomized controlled trials are required to study its effects not only in treating retinal diseases in humans but in their prevention too.
Collapse
|
20
|
Ha Y, Ochoa LF, Solomon O, Shi S, Villarreal PP, Li S, Buscho S, Vargas G, Zhang W. Light-Sheet Microscopy of the Optic Nerve Reveals Axonal Degeneration and Microglial Activation in NMDA-Induced Retinal Injury. EC OPHTHALMOLOGY 2021; 12:23-31. [PMID: 36108311 PMCID: PMC9450914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
PURPOSE Optic nerve degeneration is a feature of neurodegenerative eye diseases and causes irreversible vision loss. Therefore, understanding the degenerating patterns of the optic nerve is critical to find the potential therapeutic target for optic neuropathy. However, the traditional method of optic nerve degeneration has the limitations of losing spatiotemporal tissue information. Light sheet fluorescence microscopy (LSFM) is a fluorescence microscopy technique that allows capturing 3D images rapidly with a high spatial optical resolution. In this study, we evaluated the availability of LSFM on the optic nerve with NMDA injected Thy1-CFP mice. METHODS NMDA injected to both eyes of Thy1-CFP mice. After 7 days from the injection, the retina and optic nerve were collected and immunostained with anti-Iba1 antibody. NMDA excitotoxicity induced RGC, and its axon loss and microglial activation in the retina were observed using confocal microscopy. The immunostained optic nerve was completed the optical clearing process with TDE and mounted for LSFM imaging. RESULTS We found that retinal flatmounts confirmed significant loss of CFP-expressing RGC and axon degradation and loss in Thy1-CFP mice at 7 days after NMDA injection. Together with these data verifying that NMDA induces RGC and its axon loss, we confirmed that NMDA excitotoxicity induced microglia activation and leukostasis, such as increased microglia number, transform its morphology to ameboid or round, and increase in attached leukocytes in vessels. Using LSFM, we observed that CFP expressing nerve fiber was well organized and arranged parallel in vehicle treated optic nerve, whileas NMDA injected optic nerve showed axon swelling and fragmentation and loss of axon density from the anterior to the posterior regions. Furthermore, LSFM enabled the observation of microglia phenotype transformation in the entire optic nerve. Unlike microglia in vehicle injected optic nerve, microglia in NMDA injected optic nerve displayed larger soma and short process with high Iba1 expression through the entire optic nerve from the anterior to posterior. CONCLUSIONS In summary, we examined the applicability of the modified optic clearing protocol for the optic nerve and verified it enabled to acquiring of the 3D images of the optic nerve successfully revealing the complex spatial relationships between the axons, microglia and vasculature throughout the entire organ with single acquisitions. With these optimized techniques, we successfully obtained the high-resolution 3D images of NMDA-induced optic neuropathy, including the clues for optic nerve degeneration such as axon swelling, axonal fragmentation, and microglia activation. Overall, we believe that our current study could help understand the pathology of the optic nerve in neurodegenerative diseases, and it will be the basis for translational research.
Collapse
Affiliation(s)
- Yonju Ha
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas, USA
| | - Lorenzo F Ochoa
- Department of Neuroscience, Cell Biology and Anatomy, University of Texas Medical Branch, Galveston, Texas, USA
| | - Olivia Solomon
- Department of Neuroscience, Cell Biology and Anatomy, University of Texas Medical Branch, Galveston, Texas, USA
- Human Pathophysiology and Translational Medicine Graduate Program, University of Texas Medical Branch, Galveston, Texas, USA
| | - Shuizhen Shi
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas, USA
| | - Paula P Villarreal
- Department of Neuroscience, Cell Biology and Anatomy, University of Texas Medical Branch, Galveston, Texas, USA
| | - Shengguo Li
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas, USA
| | - Seth Buscho
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas, USA
| | - Gracie Vargas
- Department of Neuroscience, Cell Biology and Anatomy, University of Texas Medical Branch, Galveston, Texas, USA
| | - Wenbo Zhang
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas, USA
- Department of Neuroscience, Cell Biology and Anatomy, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
21
|
Kunimi H, Lee D, Ibuki M, Katada Y, Negishi K, Tsubota K, Kurihara T. Inhibition of the HIF-1α/BNIP3 pathway has a retinal neuroprotective effect. FASEB J 2021; 35:e21829. [PMID: 34314069 DOI: 10.1096/fj.202100572r] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 07/01/2021] [Accepted: 07/16/2021] [Indexed: 12/15/2022]
Abstract
Retinal ischemia is a leading cause of irreversible blindness worldwide. Inner retinal dysfunction including loss of retinal ganglion cells is encountered in a number of retinal ischemic disorders. We previously reported administration of two different hypoxia-inducible factor (HIF) inhibitors exerted neuroprotective effects in a murine model of retinal ischemia/reperfusion (I/R) which mimics these disorders, as inner retinal degeneration could be involved in pathological HIF induction. However, this notion needs further investigation. Therefore, in this study, we attempted to use retina-specific Hif-1α conditional knockout (cKO) mice to uncover this notion more clearly under the same condition. Hif-1α cKO mice showed inner retinal neurodegeneration to a lesser extent than control mice. Hif-1α depletion in a murine 661W retinal cell line reduced cell death under pseudohypoxic and hypoxic conditions. Among hypoxia-related genes, the expression of BCL2 19 kDa protein-interacting protein 3 (Bnip3) was substantially upregulated in the inner retinal layer after retinal I/R. In this regard, we further examined Bnip3 depletion in retinal neurons in vitro and in vivo and found the similar neuroprotective effects. Our results support the notion that the HIF-1α/BNIP3 pathway may have a critical role in inner retinal neurodegeneration, which can be linked with the development of new promising therapeutics for inner retinal ischemic disorders.
Collapse
Affiliation(s)
- Hiromitsu Kunimi
- Laboratory of Photobiology, Keio University School of Medicine, Shinanomachi, Japan.,Department of Ophthalmology, Keio University School of Medicine, Shinanomachi, Japan
| | - Deokho Lee
- Laboratory of Photobiology, Keio University School of Medicine, Shinanomachi, Japan.,Department of Ophthalmology, Keio University School of Medicine, Shinanomachi, Japan
| | - Mari Ibuki
- Laboratory of Photobiology, Keio University School of Medicine, Shinanomachi, Japan.,Department of Ophthalmology, Keio University School of Medicine, Shinanomachi, Japan
| | - Yusaku Katada
- Laboratory of Photobiology, Keio University School of Medicine, Shinanomachi, Japan.,Department of Ophthalmology, Keio University School of Medicine, Shinanomachi, Japan
| | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, Shinanomachi, Japan
| | - Kazuo Tsubota
- Department of Ophthalmology, Keio University School of Medicine, Shinanomachi, Japan.,Tsubota Laboratory, Inc., Tokyo, Japan
| | - Toshihide Kurihara
- Laboratory of Photobiology, Keio University School of Medicine, Shinanomachi, Japan.,Department of Ophthalmology, Keio University School of Medicine, Shinanomachi, Japan
| |
Collapse
|
22
|
Deng Y, Fu Y, Sheng L, Hu Y, Su L, Luo J, Yan C, Chi W. The Regulatory NOD-Like Receptor NLRC5 Promotes Ganglion Cell Death in Ischemic Retinopathy by Inducing Microglial Pyroptosis. Front Cell Dev Biol 2021; 9:669696. [PMID: 34095138 PMCID: PMC8173141 DOI: 10.3389/fcell.2021.669696] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
Retinal ischemia is a common pathological event that can result in retinal ganglion cell (RGC) death and irreversible vision loss. The pathogenic mechanisms linking retinal ischemia to RGC loss and visual deficits are uncertain, which has greatly hampered the development of effective treatments. It is increasingly recognized that pyroptosis of microglia contributes to the indirect inflammatory death of RGCs. In this study, we report a regulatory NOD-like receptor, NOD-, LRR- and CARD-containing 5 (NLRC5), as a key regulator on microglial pyroptosis and the retinal ischemia process. Through an in-depth analysis of our recently published transcriptome data, we found that NLRC5 was significantly up-regulated in retina during ischemia–reperfusion injury, which were further confirmed by subsequent detection of mRNA and protein level. We further found that NLRC5 was upregulated in retinal microglia during ischemia, while NLRC5 knockdown significantly ameliorated retinal ischemic damage and RGC death. Mechanistically, we revealed that knockdown of NLRC5 markedly suppressed gasdermin D (GSDMD) cleavage and activation of interleukin-1β (IL-1β) and caspase-3, indicating that NLRC5 promotes both microglial pyroptosis and apoptosis. Notably, we found that NLRC5 directly bound to NLRP3 and NLRC4 in inflammasomes to cooperatively drive microglial pyroptosis and apoptosis mediating retinal ischemic damage. Overall, these findings reveal a previously unidentified key contribution of NLRC5 signaling to microglial pyroptosis under ischemia or hypoxia conditions. This NLRC5-dependent pathway may be a novel therapeutic target for treatment of ischemic retinopathy.
Collapse
Affiliation(s)
- Yang Deng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yunzhao Fu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Longxiang Sheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yixin Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Lishi Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jiawen Luo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Chun Yan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Wei Chi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
23
|
Jiang K, Fairless E, Kanda A, Gotoh N, Cogliati T, Li T, Swaroop A. Divergent Effects of HSP70 Overexpression in Photoreceptors During Inherited Retinal Degeneration. Invest Ophthalmol Vis Sci 2021; 61:25. [PMID: 33107904 PMCID: PMC7594617 DOI: 10.1167/iovs.61.12.25] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Purpose Disruption of proteostasis is a key event in many neurodegenerative diseases. Heat shock proteins (HSPs) participate in multiple functions associated with intracellular transport and proteostasis. We evaluated the effect of augmented HSP70 expression in mutant photoreceptors of mouse retinal degeneration models to test the hypothesis that failure to sustain HSP70 expression contributes to photoreceptor cell death. Methods We examined HSP70 expression in retinas of wild-type and mutant mice by RNA and protein analysis. A transgenic mouse line, TgCrx-Hspa1a-Flag, was generated to express FLAG-tagged full-length HSP70 protein under control of a 2.3 kb mouse Crx promoter. This line was crossed to three distinct retinal degeneration mouse models. Retinal structure and function were evaluated by histology, immunohistochemistry, and electroretinography. Results In seven different mouse models of retinal degeneration, we detected transient elevation of endogenous HSP70 expression at early stages, followed by a dramatic reduction as cell death ensues, suggesting an initial adaptive response to cellular stress. Augmented expression of HSP70 in RHOT17M mice, in which mutant rhodopsin is misfolded, marginally improved photoreceptor survival, whereas elevated HSP70 led to more severe retinal degeneration in rd10 mutants that produce a partially functional PDE6B. In Rpgrip1−/− mice that display a ciliary defect, higher HSP70 had no impact on photoreceptor survival or function. Conclusions HSP70 overexpression has divergent effects in photoreceptors determined, at least in part, by the nature of the mutant protein each model carries. Additional investigations on HSP pathways and associated chaperone networks in photoreceptors are needed before designing therapeutic strategies targeting proteostasis.
Collapse
Affiliation(s)
- Ke Jiang
- Neurobiology, Neurodegeneration, and Repair Laboratory (NNRL), National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Elizabeth Fairless
- Neurobiology, Neurodegeneration, and Repair Laboratory (NNRL), National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Atsuhiro Kanda
- Neurobiology, Neurodegeneration, and Repair Laboratory (NNRL), National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Norimoto Gotoh
- Neurobiology, Neurodegeneration, and Repair Laboratory (NNRL), National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Tiziana Cogliati
- Neurobiology, Neurodegeneration, and Repair Laboratory (NNRL), National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Tiansen Li
- Neurobiology, Neurodegeneration, and Repair Laboratory (NNRL), National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Anand Swaroop
- Neurobiology, Neurodegeneration, and Repair Laboratory (NNRL), National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
24
|
Xia F, Ha Y, Shi S, Li Y, Li S, Luisi J, Kayed R, Motamedi M, Liu H, Zhang W. Early alterations of neurovascular unit in the retina in mouse models of tauopathy. Acta Neuropathol Commun 2021; 9:51. [PMID: 33762004 PMCID: PMC7992935 DOI: 10.1186/s40478-021-01149-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 03/06/2021] [Indexed: 02/07/2023] Open
Abstract
The retina, as the only visually accessible tissue in the central nervous system, has attracted significant attention for evaluating it as a biomarker for neurodegenerative diseases. Yet, most of studies focus on characterizing the loss of retinal ganglion cells (RGCs) and degeneration of their axons. There is no integrated analysis addressing temporal alterations of different retinal cells in the neurovascular unit (NVU) in particular retinal vessels. Here we assessed NVU changes in two mouse models of tauopathy, P301S and P301L transgenic mice overexpressing the human tau mutated gene, and evaluated the therapeutic effects of a tau oligomer monoclonal antibody (TOMA). We found that retinal edema and breakdown of blood-retina barrier were observed at the very early stage of tauopathy. Leukocyte adhesion/infiltration, and microglial recruitment/activation were constantly increased in the retinal ganglion cell layer of tau transgenic mice at different ages, while Müller cell gliosis was only detected in relatively older tau mice. Concomitantly, the number and function of RGCs progressively decreased during aging although they were not considerably altered in the very early stage of tauopathy. Moreover, intrinsically photosensitive RGCs appeared more sensitive to tauopathy. Remarkably, TOMA treatment in young tau transgenic mice significantly attenuated vascular leakage, inflammation and RGC loss. Our data provide compelling evidence that abnormal tau accumulation can lead to pathology in the retinal NVU, and vascular alterations occur more manifest and earlier than neurodegeneration in the retina. Oligomeric tau-targeted immunotherapy has the potential to treat tau-induced retinopathies. These data suggest that retinal NVU may serve as a potential biomarker for diagnosis and staging of tauopathy as well as a platform to study the molecular mechanisms of neurodegeneration.
Collapse
Affiliation(s)
- Fan Xia
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0144 USA
| | - Yonju Ha
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0144 USA
| | - Shuizhen Shi
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0144 USA
| | - Yi Li
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0144 USA
| | - Shengguo Li
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0144 USA
| | - Jonathan Luisi
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0144 USA
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX USA
| | - Rakez Kayed
- Department of Neurology, University of Texas Medical Branch, Galveston, TX USA
| | - Massoud Motamedi
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0144 USA
| | - Hua Liu
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0144 USA
| | - Wenbo Zhang
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0144 USA
- Departments of Neuroscience, Cell Biology and Anatomy, University of Texas Medical Branch, Galveston, TX USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555 USA
| |
Collapse
|
25
|
Liu W, Ha Y, Xia F, Zhu S, Li Y, Shi S, Mei FC, Merkley K, Vizzeri G, Motamedi M, Cheng X, Liu H, Zhang W. Neuronal Epac1 mediates retinal neurodegeneration in mouse models of ocular hypertension. J Exp Med 2020; 217:133574. [PMID: 31918438 PMCID: PMC7144517 DOI: 10.1084/jem.20190930] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 09/06/2019] [Accepted: 11/26/2019] [Indexed: 12/24/2022] Open
Abstract
Progressive loss of retinal ganglion cells (RGCs) leads to irreversible visual deficits in glaucoma. Here, we found that the level of cyclic AMP and the activity and expression of its mediator Epac1 were increased in retinas of two mouse models of ocular hypertension. Genetic depletion of Epac1 significantly attenuated ocular hypertension–induced detrimental effects in the retina, including vascular inflammation, neuronal apoptosis and necroptosis, thinning of ganglion cell complex layer, RGC loss, and retinal neuronal dysfunction. With bone marrow transplantation and various Epac1 conditional knockout mice, we further demonstrated that Epac1 in retinal neuronal cells (especially RGCs) was responsible for their death. Consistently, pharmacologic inhibition of Epac activity prevented RGC loss. Moreover, in vitro study on primary RGCs showed that Epac1 activation was sufficient to induce RGC death, which was mechanistically mediated by CaMKII activation. Taken together, these findings indicate that neuronal Epac1 plays a critical role in retinal neurodegeneration and suggest that Epac1 could be considered a target for neuroprotection in glaucoma.
Collapse
Affiliation(s)
- Wei Liu
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX.,Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yonju Ha
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX
| | - Fan Xia
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX
| | - Shuang Zhu
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX
| | - Yi Li
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX
| | - Shuizhen Shi
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX
| | - Fang C Mei
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center at Houston, Houston, TX
| | - Kevin Merkley
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX
| | - Gianmarco Vizzeri
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX
| | - Massoud Motamedi
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center at Houston, Houston, TX
| | - Hua Liu
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX
| | - Wenbo Zhang
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX.,Departments of Neuroscience, Cell Biology & Anatomy, University of Texas Medical Branch, Galveston, TX
| |
Collapse
|
26
|
Jiang N, Li Z, Li Z, Zhang Y, Yu Z, Wan P, Zhu Y, Li Y, Su W, Zhuo Y. Laquinimod exerts anti-inflammatory and antiapoptotic effects in retinal ischemia/reperfusion injury. Int Immunopharmacol 2020; 88:106989. [PMID: 33182069 DOI: 10.1016/j.intimp.2020.106989] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/22/2020] [Accepted: 09/07/2020] [Indexed: 11/15/2022]
Abstract
Retinal ischemia/reperfusion (I/R) occurs in various vision disabled ocular diseases, involved in acute glaucoma, diabetic retinopathy, ischemic optic neuropathy, hypertensive retinopathy and retinal vascular occlusion. Laquinimod (LQ), a new type of immunosuppressant, has been reported to exert anti-inflammatory effects on autoimmune diseases. This research aims to investigate the protective effect of LQ on I/R damage by focusing on inhibiting dysregulated neuroinflammation and neuronal apoptosis. In our study, mice were treated with LQ after high intraocular pressure (IOP)-induced retinal I/R injury. The data showed that LQ significantly attenuated high IOP-induced retinal ganglion cell (RGC) death and inner plexiform layer (IPL) thinning and inhibited microglial activation. The results of qRT-PCR, flow cytometry and Luminex multiplex assays demonstrated the anti-inflammatory action of LQ in BV2 cells stimulated with lipopolysaccharide (LPS). In addition, primary RGC apoptosis induced by oxygen-glucose deprivation/reperfusion (OGD/R) was also directly suppressed by LQ. Importantly, LQ inhibited the expression of cleaved caspase-8 and the downstream NLRP3 inflammasome and IL-1β. In conclusion, our findings offer the first evidence that LQ treatment prevents retinal I/R damage. Furthermore, LQ could directly inhibit RGC apoptosis. Caspase-8 activation and subsequent inflammation can also be suppressed by LQ, which suggests that LQ may act through inhibiting the caspase-8 pathway. This study demonstrates a new mechanism of LQ and provides beneficial preclinical data for the clinical application of LQ.
Collapse
Affiliation(s)
- Nan Jiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Zhidong Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Zuohong Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yingying Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Ziyu Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Peixing Wan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yingting Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yiqing Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Wenru Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| | - Yehong Zhuo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
27
|
Resveratrol protects retinal ganglion cell axons through regulation of the SIRT1-JNK pathway. Exp Eye Res 2020; 200:108249. [PMID: 32956685 DOI: 10.1016/j.exer.2020.108249] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 08/12/2020] [Accepted: 09/14/2020] [Indexed: 01/21/2023]
Abstract
It is reported that Ischemia and reperfusion damage (I/R damage) can lead to retinal ganglion cell (RGC) death and neurodegeneration, which in turn can lead to irreversible vision loss. In this study, we sought to understand the neuroprotective effect of resveratrol, the important activator of sirtuin1 (SIRT1), on RGC survival in I/R damage model and the molecular mechanism that mediate this effect. Our results show that resveratrol could reverse axonal swelling, holes, and the chaos of the nucleus in axons of RGCs caused by I/R. At the same time, resveratrol could also reverse the activation of retinal astrocytes and the loss of RGCs caused by I/R. Resveratrol increased the expression of SIRT1 while decreasing the phosphorylation of N-terminal kinase (JNK). SP600125(JNK inhibitor) decreased the phosphorylation of JNK while increasing the expression of SIRT1, indicating that SIRT1 and JNK can interact with each other. Simultaneous administration of resveratrol and sirtinol (SIRT1 inhibitor) neither increased the expression of SIRT1 nor decreased the phosphorylation of JNK, indicating that resveratrol affects the phosphorylation of JNK by SIRT1. In total, our research shows that resveratrol treatment significantly reduces apoptosis and axonal degeneration of RGCs, and this protection is partly mediated through the SIRT1-JNK pathway.
Collapse
|
28
|
Devi K, Singh N, Jaggi AS. Dual role of sirtuin 1 in inflammatory bowel disease. Immunopharmacol Immunotoxicol 2020; 42:385-391. [PMID: 32619377 DOI: 10.1080/08923973.2020.1790595] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Silent information regulator-1 (SIRT-1), is a member of the class III group of histone deacetylases and is collectively called sirtuins. There have been preclinical and clinical studies indicating the downregulation and decreased activity of sirtuin 1 in various inflammatory bowel disease models. Furthermore, the downregulation of sirtuin 1 is responsible for the sustained production of proinflammatory cytokines and the generation of oxidative stress in colitis. Hyperacetylation of NF-κB and HSF-1 (heat shock factor-1) in the absence of sirtuin1 is responsible for the induction of colitis. Accordingly, exogenous administration of sirtuin1 activators has been shown to attenuate the colitis in various inflammatory bowel disease models. On the other hand, the knockdown of sirtuin 1 gene or pharmacologic inhibition of sirtuin 1 has also been shown to be protective in the colitis. The deletion of the sirtuin1 gene may be helpful in the improvement of the disease condition of colitis through the maintenance of gastrointestinal immune homeostasis. The current review highlights the dual role of sirtuin 1 in the different experimental models of IBD along with possible mechanisms.
Collapse
Affiliation(s)
- Karam Devi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Nirmal Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Amteshwar Singh Jaggi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| |
Collapse
|
29
|
López-Malo D, Villarón-Casares CA, Alarcón-Jiménez J, Miranda M, Díaz-Llopis M, Romero FJ, Villar VM. Curcumin as a Therapeutic Option in Retinal Diseases. Antioxidants (Basel) 2020; 9:antiox9010048. [PMID: 31935797 PMCID: PMC7023263 DOI: 10.3390/antiox9010048] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/27/2019] [Accepted: 12/29/2019] [Indexed: 12/18/2022] Open
Abstract
The retina is subjected to oxidative stress due to its high vascularization, long time light exposition and a high density of mitochondria. Oxidative stress can lead to pathological processes, like cell apoptosis, angiogenesis and inflammation ending in retinal pathologies. Curcumin, a major bioactive component obtained from the spice turmeric (Curcuma longa) rhizome has been used for centuries in Asian countries for cooking and for curing all kinds of diseases like dysentery, chest congestion and pain in general, due to its antioxidant effects. Curcumin prevents the formation of reactive oxygen species and so it is a good protective agent. Curcumin has shown also anti-inflammatory, and antitumor properties. Curcumin is a natural product, which can be a therapeutic option in a variety of retinal diseases due to its pleiotropic properties. Some drawbacks are its poor solubility, bioavailability and lack of stability at physiological conditions; which have been shown in curcumin skeptical publications. In this review, we provide some lights and shadows on curcumin administration on the major retinal pathologies.
Collapse
Affiliation(s)
- Daniel López-Malo
- Facultad de Ciencias de la Salud, Universidad Europea de Valencia, 46010 Valencia, Spain; (D.L.-M.); (C.A.V.-C.)
| | | | - Jorge Alarcón-Jiménez
- Facultad de Medicina y Ciencias de la Salud, Universidad Católica de Valencia, 46001 Valencia, Spain;
| | - Maria Miranda
- Facultad de Ciencias de la Salud, Universidad CEU Cardenal Herrera, 46315 Moncada, Spain; (M.M.); (V.M.V.)
| | - Manuel Díaz-Llopis
- Facultad de Medicina y Odontología, Universitat de Valencia, 46010 Valencia, Spain;
| | - Francisco J. Romero
- Facultad de Ciencias de la Salud, Universidad Europea de Valencia, 46010 Valencia, Spain; (D.L.-M.); (C.A.V.-C.)
- Hospital General de Requena, Conselleria de Sanitat, Generalitat Valenciana, 46340 Valencia, Spain
- Correspondence: ; Tel.: +34-961-0438-83
| | - Vincent M. Villar
- Facultad de Ciencias de la Salud, Universidad CEU Cardenal Herrera, 46315 Moncada, Spain; (M.M.); (V.M.V.)
| |
Collapse
|
30
|
Kunimi H, Miwa Y, Katada Y, Tsubota K, Kurihara T. HIF inhibitor topotecan has a neuroprotective effect in a murine retinal ischemia-reperfusion model. PeerJ 2019; 7:e7849. [PMID: 31592359 PMCID: PMC6779112 DOI: 10.7717/peerj.7849] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 09/08/2019] [Indexed: 12/13/2022] Open
Abstract
Purpose The therapeutic approach for retinal ganglion cell (RGC) degeneration has not been fully established. Recently, it has been reported that hypoxia-inducible factor (HIF) may be involved with retinal neurodegeneration. In this study, we investigated neuroprotective effects of a HIF inhibitor against RGC degeneration induced in a murine model of retinal ischemia-reperfusion (I/R). Methods Eight-weeks-old male C57/BL6J mice were treated with intraperitoneal injection of a HIF inhibitor topotecan (1.25 mg/kg) for 14 days followed by a retinal I/R procedure. Seven days after the I/R injury, the therapeutic effect was evaluated histologically and electrophysiologically. Results The increase of HIF-1α expression and the decrease of retinal thickness and RGC number in I/R were significantly suppressed by administration of topotecan. Impaired visual function in I/R was improved by topotecan evaluated with electroretinogram and visual evoked potentials. Conclusions Topotecan administration suppressed HIF-1a expression and improved RGC survival resulting in a functional protection against retinal I/R. These data indicated that the HIF inhibitor topotecan may have therapeutic potentials for RGC degeneration induced with retinal ischemia or high intraocular pressure.
Collapse
Affiliation(s)
- Hiromitsu Kunimi
- Department of Ophthalmology, School of Medicine, Keio University, Tokyo, Japan.,Laboratory of Photobiology, School of Medicine, Keio University, Tokyo, Japan
| | - Yukihiro Miwa
- Department of Ophthalmology, School of Medicine, Keio University, Tokyo, Japan.,Laboratory of Photobiology, School of Medicine, Keio University, Tokyo, Japan
| | - Yusaku Katada
- Department of Ophthalmology, School of Medicine, Keio University, Tokyo, Japan.,Laboratory of Photobiology, School of Medicine, Keio University, Tokyo, Japan
| | - Kazuo Tsubota
- Department of Ophthalmology, School of Medicine, Keio University, Tokyo, Japan
| | - Toshihide Kurihara
- Department of Ophthalmology, School of Medicine, Keio University, Tokyo, Japan.,Laboratory of Photobiology, School of Medicine, Keio University, Tokyo, Japan
| |
Collapse
|
31
|
Coucha M, Shanab AY, Sayed M, Vazdarjanova A, El-Remessy AB. Modulating Expression of Thioredoxin Interacting Protein (TXNIP) Prevents Secondary Damage and Preserves Visual Function in a Mouse Model of Ischemia/Reperfusion. Int J Mol Sci 2019; 20:ijms20163969. [PMID: 31443163 PMCID: PMC6721134 DOI: 10.3390/ijms20163969] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 12/29/2022] Open
Abstract
Retinal neurodegeneration, an early characteristic of several blinding diseases, triggers glial activation, resulting in inflammation, secondary damage and visual impairment. Treatments that aim only at neuroprotection have failed clinically. Here, we examine the impact of modulating thioredoxin interacting protein (TXNIP) to the inflammatory secondary damage and visual impairment in a model of ischemia/reperfusion (IR). Wild type (WT) and TXNIP knockout (TKO) mice underwent IR injury by increasing intraocular pressure for 40 min, followed by reperfusion. An additional group of WT mice received intravitreal TXNIP-antisense oligomers (ASO, 100 µg/2 µL) 2 days post IR injury. Activation of Müller glial cells, apoptosis and expression of inflammasome markers and visual function were assessed. IR injury triggered early TXNIP mRNA expression that persisted for 14 days and was localized within activated Müller cells in WT-IR, compared to sham controls. Exposure of Müller cells to hypoxia-reoxygenation injury triggered endoplasmic reticulum (ER) stress markers and inflammasome activation in WT cells, but not from TKO cells. Secondary damage was evident by the significant increase in the number of occluded acellular capillaries and visual impairment in IR-WT mice but not in IR-TKO. Intervention with TXNIP-ASO prevented ischemia-induced glial activation and neuro-vascular degeneration, and improved visual function compared to untreated WT. Targeting TXNIP expression may offer an effective approach in the prevention of secondary damage associated with retinal neurodegenerative diseases.
Collapse
Affiliation(s)
- Maha Coucha
- Augusta Biomedical Research Corporation, Augusta, GA 30901, USA
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
- Department of Pharmaceutical Sciences, South University, School of Pharmacy, Savannah, GA 31406, USA
| | - Ahmed Y Shanab
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| | - Mohamed Sayed
- Augusta Biomedical Research Corporation, Augusta, GA 30901, USA
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| | - Almira Vazdarjanova
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
- Department of Pharmacology and Toxicology, Augusta University, Augusta, GA 30901, USA
| | - Azza B El-Remessy
- Augusta Biomedical Research Corporation, Augusta, GA 30901, USA.
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA.
- Department of Pharmacy, Doctors Hospital of Augusta, Augusta, GA 30909, USA.
| |
Collapse
|
32
|
The Anti-Inflammatory Effects of CXCR5 in the Mice Retina following Ischemia-Reperfusion Injury. BIOMED RESEARCH INTERNATIONAL 2019; 2019:3487607. [PMID: 31355256 PMCID: PMC6637708 DOI: 10.1155/2019/3487607] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 04/22/2019] [Accepted: 04/30/2019] [Indexed: 01/18/2023]
Abstract
Object Retinal ischemia-reperfusion (I/R) injury is a common pathological process in many ophthalmic diseases; there are no effective therapeutic approaches available currently. Increasing evidence indicates that microglia mediated neuroinflammation plays an important role in the retinal I/R injury. In this study, we aimed to investigate the roles of chemokine receptor CXCR5 in the pathological process of retinal I/R injury model. Method Retinal I/R injury model was established in CXCR5 knockout and wild mice by the acute elevation of intraocular pressure (AOH) for 60 minutes, and the eyes were harvested for further analyses. The cellular location of CXCR5 was detected by immunofluorescence staining; the expressions of CXCR5 and CXCL13 after I/R injury were analyzed by quantitative RT-PCR. The retinal microglia were detected as stained for Iba1 (+). Leakage of inflammatory cells was observed on the H&E stained cryosections. The protein expression and quantification of zonula occludens (ZO-1) were determined by Western blotting and densitometry. Capillary degeneration was identified on the intact retinal vasculatures prepared by trypsin digestion. Results The number of activated microglia marked by Iba1 antibody in the retina was increased after retinal I/R injury in both KO and WT mice, more significant in KO mice. The leakage of inflammatory cells was observed largely at 2 days after injury, but there was no or little leakage at 7 days. The number of inflammatory cells (mainly neutrophils) was greater in CXCR5 KO mice than in WT mice, mainly located under internal limiting membrane. CXCR5 deficiency led to more ZO-1 degradation in CXCR5 KO mice compared to C57BL6 WT mice 2 days after reperfusion. The cellular capillaries were also significantly increased in the KO mice compared to the WT mice. Conclusion Our findings suggest that the chemokine receptor CXCR5 may protect retina from ischemia-reperfusion injury by its anti-inflammatory effects. Thus, CXCR5 may be a promising therapeutic target for the treatment of retinal I/R injury.
Collapse
|