1
|
Scalici A, Miller-Fleming TW, Shuey MM, Baker JT, Betti M, Hirbo J, Knapik EW, Cox NJ. Gene and phenome-based analysis of the shared genetic architecture of eye diseases. Am J Hum Genet 2025; 112:318-331. [PMID: 39879988 PMCID: PMC11866973 DOI: 10.1016/j.ajhg.2025.01.004] [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: 06/21/2024] [Revised: 12/31/2024] [Accepted: 01/03/2025] [Indexed: 01/31/2025] Open
Abstract
While many eye disorders are linked through defects in vascularization and optic nerve degeneration, genetic correlation studies have yielded variable results despite shared features. For example, glaucoma and myopia both share optic neuropathy as a feature, but genetic correlation studies demonstrated minimal overlap. By leveraging electronic health record (EHR) resources that contain genetic variables such as genetically predicted gene expression (GPGE), researchers have the potential to improve the identification of shared genetic drivers of disease by incorporating knowledge of shared features to identify disease-causing mechanisms. In this study, we examined shared genetic architecture across eye diseases. Our gene-based approach used transcriptome-wide association methods to identify shared transcriptomic profiles across eye diseases within BioVU, Vanderbilt University Medical Center's (VUMC's) EHR-linked biobank. Our phenome-based approach leveraged phenome-wide association studies (PheWASs) to identify eye disease comorbidities. Using the beta estimates from the significantly associated comorbidities, we constructed a phenotypic risk score (PheRS) representing a weighted sum of an individual's eye disease comorbidities. This PheRS is predictive of eye disease status and associated with the altered GPGE of significant genes in an independent population. The implementation of both gene- and phenome-based approaches can expand genetic associations and shed greater insight into the underlying mechanisms of shared genetic architecture across eye diseases.
Collapse
Affiliation(s)
- Alexandra Scalici
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA; Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tyne W Miller-Fleming
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA; Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Megan M Shuey
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA; Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - James T Baker
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA; Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael Betti
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA; Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jibril Hirbo
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA; Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ela W Knapik
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA; Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nancy J Cox
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA; Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
2
|
Nam MH, Dhillon A, Nahomi RB, Carrillo NL, Hougen CS, Nagaraj RH. Peptain-1 blocks ischemia/reperfusion-induced retinal capillary degeneration in mice. Front Cell Neurosci 2024; 18:1441924. [PMID: 39149168 PMCID: PMC11324586 DOI: 10.3389/fncel.2024.1441924] [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: 05/31/2024] [Accepted: 07/16/2024] [Indexed: 08/17/2024] Open
Abstract
Introduction Neurovascular degeneration results in vascular dysfunction, leakage, ischemia, and structural changes that can lead to significant visual impairment. We previously showed the protective effects of peptain-1, a 20 amino acid peptide derived from the αB-crystallin core domain, on retinal ganglion cells in two animal models of glaucoma. Here, we evaluated the ability of peptain-1 to block apoptosis of human retinal endothelial cells (HRECs) in vitro and retinal capillary degeneration in mice subjected to retinal ischemia/reperfusion (I/R) injury. Methods HRECs were treated with either peptain-1 or scrambled peptides (200 μg/mL) for 3 h and a combination of proinflammatory cytokines (IFN-γ 20 ng/mL + TNF-α 20 ng/mL+ IL-1β 20 ng/mL) for additional 48 h. Apoptosis was measured with cleaved caspase-3 formation via western blot, and by TUNEL assay. C57BL/6J mice (12 weeks old) were subjected to I/R injury by elevating the intraocular pressure to 120 mmHg for 60 min, followed by reperfusion. Peptain-1 or scrambled peptide (0.5 μg) was intravitreally injected immediately after I/R injury and 7 days later. One microliter of PBS was injected as vehicle control, and animals were euthanized on day 14 post-I/R injury. Retinal capillary degeneration was assessed after enzyme digestion followed by periodic acid-Schiff staining. Results Our data showed that peptain-1 entered HRECs and blocked proinflammatory cytokine-mediated apoptosis. Intravitreally administered peptain-1 was distributed throughout the retinal vessels after 4 h. I/R injury caused retinal capillary degeneration. Unlike scrambled peptide, peptain-1 protected capillaries against I/R injury. Additionally, peptain-1 inhibited microglial activation and reduced proinflammatory cytokine levels in the retina following I/R injury. Discussion Our study suggests that peptain-1 could be used as a therapeutic agent to prevent capillary degeneration and neuroinflammation in retinal ischemia.
Collapse
Affiliation(s)
- Mi-Hyun Nam
- Department of Ophthalmology, UCHealth-Sue Anschutz-Rodgers Eye Centre, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Armaan Dhillon
- Department of Ophthalmology, UCHealth-Sue Anschutz-Rodgers Eye Centre, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Rooban B Nahomi
- Department of Ophthalmology, UCHealth-Sue Anschutz-Rodgers Eye Centre, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Noelle L Carrillo
- Department of Ophthalmology, UCHealth-Sue Anschutz-Rodgers Eye Centre, School of Medicine, University of Colorado, Aurora, CO, United States
- Department of Radiology, UCHealth University of Colorado Hospital, Aurora, CO, United States
| | - Clarinda S Hougen
- Department of Ophthalmology, UCHealth-Sue Anschutz-Rodgers Eye Centre, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Ram H Nagaraj
- Department of Ophthalmology, UCHealth-Sue Anschutz-Rodgers Eye Centre, School of Medicine, University of Colorado, Aurora, CO, United States
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, United States
| |
Collapse
|
3
|
Pang Y, Hu H, Xu K, Cao T, Wang Z, Nie J, Zheng H, Luo H, Wang F, Xiong C, Deng KY, Xin HB, Zhang X. CD38 Deficiency Protects Mouse Retinal Ganglion Cells Through Activating the NAD+/Sirt1 Pathway in Ischemia-Reperfusion and Optic Nerve Crush Models. Invest Ophthalmol Vis Sci 2024; 65:36. [PMID: 38776115 PMCID: PMC11127494 DOI: 10.1167/iovs.65.5.36] [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: 12/27/2023] [Accepted: 05/05/2024] [Indexed: 05/27/2024] Open
Abstract
Purpose The purpose of this study was to investigate the protective effect of CD38 deletion on retinal ganglion cells (RGCs) in a mouse retinal ischemia/reperfusion (I/R) model and an optic nerve crush (ONC) model, and to elucidate the underlying molecular mechanisms. Methods Retinal I/R and ONC models were constructed in mice. PCR was used to identify the deletion of CD38 gene in mice, hematoxylin and eosin (H&E) staining was used to evaluate the changes in retinal morphology, and electroretinogram (ERG) was used to evaluate the changes in retinal function. The survival of RGCs and activation of retinal macroglia were evaluated by immunofluorescence staining. The expression of Sirt1, CD38, Ac-p65, Ac-p53, TNF-α, IL-1β, and Caspase3 proteins in the retina was further evaluated by protein imprinting. Results In retinal I/R and ONC models, CD38 deficiency reduced the loss of RGCs and activation of macroglia and protected the retinal function. CD38 deficiency increased the concentration of NAD+, reduced the degree of acetylation of NF-κB p65 and p53, and reduced expression of the downstream inflammatory cytokines TNFα, IL-1β, and apoptotic protein Caspase3 in the retina in the ONC model. Intraperitoneal injection of the Sirt1 inhibitor EX-527 partially counteracted the effects of CD38 deficiency, suggesting that CD38 deficiency acts at least in part through the NAD+/Sirt1 pathway. Conclusions CD38 plays an important role in the pathogenesis of retinal I/R and ONC injury. CD38 deletion protects RGCs by attenuating inflammatory responses and apoptosis through the NAD+/Sirt1 pathway.
Collapse
Affiliation(s)
- Yulian Pang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, Nanchang, China
| | - Haijian Hu
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, Nanchang, China
| | - Ke Xu
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, Nanchang, China
| | - Ting Cao
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, Nanchang, China
- Department of Orthopaedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Zhiruo Wang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, Nanchang, China
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jiahe Nie
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, Nanchang, China
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Haina Zheng
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, Nanchang, China
| | - Hongdou Luo
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, Nanchang, China
| | - Feifei Wang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, Nanchang, China
| | - Chan Xiong
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, Nanchang, China
| | - Ke-Yu Deng
- Institute of Translational Medicine, Nanchang University, Nanchang, China
| | - Hong-Bo Xin
- Institute of Translational Medicine, Nanchang University, Nanchang, China
| | - Xu Zhang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, Nanchang, China
| |
Collapse
|
4
|
Kim YA, Choi Y, Kim TG, Jeong J, Yu S, Kim T, Sheen K, Lee Y, Choi T, Park YH, Kang MS, Kim MS. Multi-System-Level Analysis with RNA-Seq on Pterygium Inflammation Discovers Association between Inflammatory Responses, Oxidative Stress, and Oxidative Phosphorylation. Int J Mol Sci 2024; 25:4789. [PMID: 38732006 PMCID: PMC11083828 DOI: 10.3390/ijms25094789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/18/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
A pterygium is a common conjunctival degeneration and inflammatory condition. It grows onto the corneal surface or limbus, causing blurred vision and cosmetic issues. Ultraviolet is a well-known risk factor for the development of a pterygium, although its pathogenesis remains unclear, with only limited understanding of its hereditary basis. In this study, we collected RNA-seq from both pterygial tissues and conjunctival tissues (as controls) from six patients (a total of twelve biological samples) and retrieved publicly available data, including eight pterygium samples and eight controls. We investigated the intrinsic gene regulatory mechanisms closely linked to the inflammatory reactions of pterygiums and compared Asian (Korea) and the European (Germany) pterygiums using multiple analysis approaches from different perspectives. The increased expression of antioxidant genes in response to oxidative stress and DNA damage implies an association between these factors and pterygium development. Also, our comparative analysis revealed both similarities and differences between Asian and European pterygiums. The decrease in gene expressions involved in the three primary inflammatory signaling pathways-JAK/STAT, MAPK, and NF-kappa B signaling-suggests a connection between pathway dysfunction and pterygium development. We also observed relatively higher activity of autophagy and antioxidants in the Asian group, while the European group exhibited more pronounced stress responses against oxidative stress. These differences could potentially be necessitated by energy-associated pathways, specifically oxidative phosphorylation.
Collapse
Affiliation(s)
- Ye-Ah Kim
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul 05278, Republic of Korea; (Y.-A.K.); (Y.C.); (J.J.); (S.Y.); (T.K.); (K.S.); (Y.L.)
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Yueun Choi
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul 05278, Republic of Korea; (Y.-A.K.); (Y.C.); (J.J.); (S.Y.); (T.K.); (K.S.); (Y.L.)
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Tae Gi Kim
- Department of Ophthalmology, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul 05278, Republic of Korea;
| | - Jisu Jeong
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul 05278, Republic of Korea; (Y.-A.K.); (Y.C.); (J.J.); (S.Y.); (T.K.); (K.S.); (Y.L.)
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Sanghyeon Yu
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul 05278, Republic of Korea; (Y.-A.K.); (Y.C.); (J.J.); (S.Y.); (T.K.); (K.S.); (Y.L.)
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Taeyoon Kim
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul 05278, Republic of Korea; (Y.-A.K.); (Y.C.); (J.J.); (S.Y.); (T.K.); (K.S.); (Y.L.)
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Kisung Sheen
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul 05278, Republic of Korea; (Y.-A.K.); (Y.C.); (J.J.); (S.Y.); (T.K.); (K.S.); (Y.L.)
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Yoonsung Lee
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul 05278, Republic of Korea; (Y.-A.K.); (Y.C.); (J.J.); (S.Y.); (T.K.); (K.S.); (Y.L.)
| | - Taesoo Choi
- Department of Urology, School of Medicine, Kyung Hee University, Seoul 05278, Republic of Korea;
| | - Yong Hwan Park
- Department of Microbiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea;
| | - Min Seok Kang
- Department of Ophthalmology, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul 02447, Republic of Korea
| | - Man S. Kim
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul 05278, Republic of Korea; (Y.-A.K.); (Y.C.); (J.J.); (S.Y.); (T.K.); (K.S.); (Y.L.)
| |
Collapse
|
5
|
Jin Q, Liu T, Ma F, Fu T, Yang L, Mao H, Wang Y, Peng L, Li P, Zhan Y. Roles of Sirt1 and its modulators in diabetic microangiopathy: A review. Int J Biol Macromol 2024; 264:130761. [PMID: 38467213 DOI: 10.1016/j.ijbiomac.2024.130761] [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: 12/27/2023] [Revised: 02/03/2024] [Accepted: 03/07/2024] [Indexed: 03/13/2024]
Abstract
Diabetic vascular complications include diabetic macroangiopathy and diabetic microangiopathy. Diabetic microangiopathy is characterised by impaired microvascular endothelial function, basement membrane thickening, and microthrombosis, which may promote renal, ocular, cardiac, and peripheral system damage in diabetic patients. Therefore, new preventive and therapeutic strategies are urgently required. Sirt1, a member of the nicotinamide adenine dinucleotide-dependent histone deacetylase class III family, regulates different organ growth and development, oxidative stress, mitochondrial function, metabolism, inflammation, and aging. Sirt1 is downregulated in vascular injury and microangiopathy. Moreover, its expression and distribution in different organs correlate with age and play critical regulatory roles in oxidative stress and inflammation. This review introduces the background of diabetic microangiopathy and the main functions of Sirt1. Then, the relationship between Sirt1 and different diabetic microangiopathies and the regulatory roles mediated by different cells are described. Finally, we summarize the modulators that target Sirt1 to ameliorate diabetic microangiopathy as an essential preventive and therapeutic measure for diabetic microangiopathy. In conclusion, targeting Sirt1 may be a new therapeutic strategy for diabetic microangiopathy.
Collapse
Affiliation(s)
- Qi Jin
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tongtong Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fang Ma
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tongfei Fu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liping Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Huimin Mao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuyang Wang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liang Peng
- China-Japan Friendship Hospital, Institute of Clinical Medical Sciences, Beijing, China.
| | - Ping Li
- China-Japan Friendship Hospital, Institute of Clinical Medical Sciences, Beijing, China.
| | - Yongli Zhan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
6
|
Zeng L, Ying Q, Lou H, Wang F, Pang Y, Hu H, Zhang Z, Song Y, Liu P, Zhang X. Protective effect of the natural flavonoid naringenin in mouse models of retinal injury. Eur J Pharmacol 2024; 962:176231. [PMID: 38052414 DOI: 10.1016/j.ejphar.2023.176231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/26/2023] [Accepted: 11/27/2023] [Indexed: 12/07/2023]
Abstract
Glaucoma is an eye disease with a high rate of blindness and a complex pathogenesis. Ocular hypertension (OHT) is a critical risk factor, and retinal ischemia/reperfusion (I/R) is an important pathophysiological basis. This study was designed to investigate the retinal neuroprotective effect of oral naringenin in an acute retinal I/R model and a chronic OHT model and the possible mechanism involved. After the I/R and OHT models were established, mice were given vehicle or naringenin (100 mg/kg or 300 mg/kg). Hematoxylin-eosin (HE) staining and immunostaining of RBPMS and glial fibrillary acidic protein (GFAP) were used to evaluate retinal injury. GFAP, CD38, Sirtuin1 (SIRT1), and NOD-like receptor protein 3 (NLRP3) expression levels were measured by Western blotting. In the OHT model, intraocular pressure (IOP) was dynamically maintained at approximately 20-25 mmHg after injury. The retinal structure was damaged, and retinal ganglion cells (RGCs) were lost in both models. Naringenin ameliorated the abovementioned indications but also demonstrated that high concentrations of naringenin significantly inhibited retinal astrocyte activation and inhibited damage-induced increases in the expression of GFAP, NLRP3, and CD38 proteins, while SIRT1 protein expression was upregulated. This study showed for the first time that naringenin can reduce microbead-induced IOP elevation in the OHT model, providing new evidence for the application of naringenin in glaucoma. Naringenin may mediate the CD38/SIRT1 signaling pathway, inhibit astrocyte activation, and ultimately exert an anti-inflammatory effect to achieve retinal neuroprotection.
Collapse
Affiliation(s)
- Ling Zeng
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, Jiangxi, China
| | - Qian Ying
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, Jiangxi, China
| | - Hongdou Lou
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, Jiangxi, China
| | - Feifei Wang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, Jiangxi, China
| | - Yulian Pang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, Jiangxi, China
| | - Haijian Hu
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, Jiangxi, China
| | - Ziqiao Zhang
- Queen Mary School, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Yuning Song
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, Jiangxi, China
| | - Peiyu Liu
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, Jiangxi, China
| | - Xu Zhang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, Jiangxi, China.
| |
Collapse
|
7
|
Zhang Z, Peng S, Xu T, Liu J, Zhao L, Xu H, Zhang W, Zhu Y, Yang Z. Retinal Microenvironment-Protected Rhein-GFFYE Nanofibers Attenuate Retinal Ischemia-Reperfusion Injury via Inhibiting Oxidative Stress and Regulating Microglial/Macrophage M1/M2 Polarization. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302909. [PMID: 37653617 PMCID: PMC10602545 DOI: 10.1002/advs.202302909] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/20/2023] [Indexed: 09/02/2023]
Abstract
Retinal ischemia is involved in the occurrence and development of various eye diseases, including glaucoma, diabetic retinopathy, and central retinal artery occlusion. To the best of our knowledge, few studies have reported self-assembling peptide natural products for the suppression of ocular inflammation and oxidative stress. Herein, a self-assembling peptide GFFYE is designed and synthesized, which can transform the non-hydrophilicity of rhein into an amphiphilic sustained-release therapeutic agent, and rhein-based therapeutic nanofibers (abbreviated as Rh-GFFYE) are constructed for the treatment of retinal ischemia-reperfusion (RIR) injury. Rh-GFFYE significantly ameliorates oxidative stress and inflammation in an in vitro oxygen-glucose deprivation (OGD) model of retinal ischemia and a rat model of RIR injury. Rh-GFFYE also significantly enhances retinal electrophysiological recovery and exhibits good biocompatibility. Importantly, Rh-GFFYE also promotes the transition of M1-type macrophages to the M2 type, ultimately altering the pro-inflammatory microenvironment. Further investigation of the treatment mechanism indicates that Rh-GFFYE activates the PI3K/AKT/mTOR signaling pathway to reduce oxidative stress and inhibits the NF-κB and STAT3 signaling pathways to affect inflammation and macrophage polarization. In conclusion, the rhein-loaded nanoplatform alleviates RIR injury by modulating the retinal microenvironment. The findings are expected to promote the clinical application of hydrophobic natural products in RIR injury-associated eye diseases.
Collapse
Affiliation(s)
- Zhuhong Zhang
- School of PharmacyKey Laboratory of Molecular Pharmacology and Drug EvaluationMinistry of EducationCollaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of ShandongYantai UniversityYantai264005China
| | - Shengjun Peng
- School of PharmacyKey Laboratory of Molecular Pharmacology and Drug EvaluationMinistry of EducationCollaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of ShandongYantai UniversityYantai264005China
| | - Tengyan Xu
- Key Laboratory of Bioactive MaterialsMinistry of EducationState Key Laboratory of Medicinal Chemical BiologyCollege of Life SciencesCollaborative Innovation Center of Chemical Science and Engineeringand National Institute of Functional MaterialsNankai UniversityTianjin300071China
| | - Jia Liu
- School of PharmacyKey Laboratory of Molecular Pharmacology and Drug EvaluationMinistry of EducationCollaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of ShandongYantai UniversityYantai264005China
| | - Laien Zhao
- School of PharmacyKey Laboratory of Molecular Pharmacology and Drug EvaluationMinistry of EducationCollaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of ShandongYantai UniversityYantai264005China
| | - Hui Xu
- School of PharmacyKey Laboratory of Molecular Pharmacology and Drug EvaluationMinistry of EducationCollaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of ShandongYantai UniversityYantai264005China
| | - Wen Zhang
- School of PharmacyKey Laboratory of Molecular Pharmacology and Drug EvaluationMinistry of EducationCollaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of ShandongYantai UniversityYantai264005China
| | - Yuanying Zhu
- School of PharmacyKey Laboratory of Molecular Pharmacology and Drug EvaluationMinistry of EducationCollaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of ShandongYantai UniversityYantai264005China
| | - Zhimou Yang
- Key Laboratory of Bioactive MaterialsMinistry of EducationState Key Laboratory of Medicinal Chemical BiologyCollege of Life SciencesCollaborative Innovation Center of Chemical Science and Engineeringand National Institute of Functional MaterialsNankai UniversityTianjin300071China
| |
Collapse
|
8
|
Liu S, Zhang W. NAD + metabolism and eye diseases: current status and future directions. Mol Biol Rep 2023; 50:8653-8663. [PMID: 37540459 DOI: 10.1007/s11033-023-08692-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 07/18/2023] [Indexed: 08/05/2023]
Abstract
Currently, there are no truly effective treatments for a variety of eye diseases, such as glaucoma, age-related macular degeneration (AMD), and inherited retinal degenerations (IRDs). These conditions have a significant impact on patients' quality of life and can be a burden on society. However, these diseases share a common pathological process of NAD+ metabolism disorders. They are either associated with genetically induced primary NAD+ synthase deficiency, decreased NAD+ levels due to aging, or enhanced NAD+ consuming enzyme activity during disease pathology. In this discussion, we explore the role of NAD+ metabolic disorders in the development of associated ocular diseases and the potential advantages and disadvantages of various methods to increase NAD+ levels. It is essential to carefully evaluate the possible adverse effects of these methods and conduct a more comprehensive and objective assessment of their function before considering their use.
Collapse
Affiliation(s)
- Siyuan Liu
- Department of Ophthalmology, Second Clinical Medical College, Lanzhou University, 730030, Lanzhou, VA, China
| | - Wenfang Zhang
- Department of Ophthalmology, The Second Hospital of Lanzhou University, 730030, Lanzhou, VA, China.
| |
Collapse
|
9
|
Zhang Z, Wang L, Zeng D, Ma X, Wang H. Lotus-Flower- and Lotus-Seedpod-Derived Polysaccharide: Structural Characterization and Biological Activity. Polymers (Basel) 2023; 15:3828. [PMID: 37765682 PMCID: PMC10536264 DOI: 10.3390/polym15183828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/31/2023] [Accepted: 02/07/2023] [Indexed: 09/29/2023] Open
Abstract
Lotus flower polysaccharide (LFP) and lotus seedpod polysaccharide (LSP) were separated by water extract-alcohol precipitation, and their structures and biological activities were investigated. The results of monosaccharide composition showed that LFP and LSP were composed of nine monosaccharides, fucose, rhamnose, arabinose, glucose, galactose, mannose, fructose, galacturonic acid, and glucuronic acid, with the molar percentages of 0.18: 0.43: 2.26: 45.22: 32.14: 4.28: 8.20: 6.28: 1.01 and 2.70: 1.02: 8.15: 45.63: 20.63: 1.44: 2.59: 16.45. LSP and LFP exhibited molecular weights of 9.37 × 104 Da and 1.24 × 106 Da, respectively. SEM showed that LFP and LSP have similar structures; XRD analysis showed that both polysaccharides had crystalline structure and amorphous structure. The results of ABTS+, DPPH, hydroxyl radical scavenging experiment, and a reducing power experiment showed that LFP and LSP had good antioxidant capacity. Cell viability findings showed that polysaccharide concentrations of lotus flower and lotus seedpod could enhance cellular proliferation ranging from 25 to 400 μg/mL without cytotoxicity. By inducing the production of crucial proteins in the TLR4/NF-κB pathway, LFP and LSP were able to induce autophagy in RAW264.7, according to the results of the RT-PCR and Western blotting assays.
Collapse
Affiliation(s)
- Zhiqiang Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450045, China
| | - Li Wang
- Department of Traditional Chinese Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Dai Zeng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450045, China
| | - Xia Ma
- College of Animal Medicine, Henan University of Animal Husbandry and Economy, Zhengzhou 450046, China
| | - Hui Wang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450045, China
| |
Collapse
|
10
|
Wang F, Song Y, Liu P, Ma F, Peng Z, Pang Y, Hu H, Zeng L, Luo H, Zhang X. Rapamycin suppresses neuroinflammation and protects retinal ganglion cell loss after optic nerve crush. Int Immunopharmacol 2023; 119:110171. [PMID: 37060809 DOI: 10.1016/j.intimp.2023.110171] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/26/2023] [Accepted: 04/07/2023] [Indexed: 04/17/2023]
Abstract
Pyroptosis, an inflammasome-mediated mode of death, plays an important role in glaucoma. It has been shown that regulating the mTOR pathway can inhibit pyroptosis. Unfortunately, whether rapamycin (RAPA), a specific inhibitor of the mTOR pathway, can inhibit optic nerve crush (ONC)-induced pyroptosis to protect retinal ganglion cells (RGCs) has not been investigated. Our research aimed to confirm the effect of intravitreal injection of RAPA on RGCs. Furthermore, we used the ONC model to explore the underlying mechanisms. First, we observed that intravitreal injection of RAPA alleviated RGC damage induced by various types of injury. We then used the ONC model to further explore the potential mechanism of RAPA. Mechanistically, RAPA not only reduced the activation of glial cells in the retina but also inhibited retinal pyroptosis-induced expression of inflammatory factors such as nucleotide-binding oligomeric domain-like receptor 3 (NLRP3), apoptosis-associated speckle-like protein containing a CARD (ASC), N-terminal of gasdermin-D (GSDMD-N), IL-18 and IL-1β. Moreover, RAPA exerted protective effects on RGC axons, possibly by inhibiting glial activation and regulating the mTOR/ROCK pathway. Therefore, this study demonstrates a novel mechanism by which RAPA protects against glaucoma and provides further evidence for its application in preclinical studies.
Collapse
Affiliation(s)
- Feifei Wang
- Affiliated Eye Hospital of Nanchang University, Nanchang University School of Ophthalmology & Optometry, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Research Institute of Ophthalmology & Visual Science, Nanchang 330006, PR China
| | - Yuning Song
- Affiliated Eye Hospital of Nanchang University, Nanchang University School of Ophthalmology & Optometry, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Research Institute of Ophthalmology & Visual Science, Nanchang 330006, PR China
| | - Peiyu Liu
- Affiliated Eye Hospital of Nanchang University, Nanchang University School of Ophthalmology & Optometry, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Research Institute of Ophthalmology & Visual Science, Nanchang 330006, PR China
| | - Fangli Ma
- Affiliated Eye Hospital of Nanchang University, Nanchang University School of Ophthalmology & Optometry, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Research Institute of Ophthalmology & Visual Science, Nanchang 330006, PR China
| | - Zhida Peng
- Affiliated Eye Hospital of Nanchang University, Nanchang University School of Ophthalmology & Optometry, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Research Institute of Ophthalmology & Visual Science, Nanchang 330006, PR China
| | - Yulian Pang
- Affiliated Eye Hospital of Nanchang University, Nanchang University School of Ophthalmology & Optometry, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Research Institute of Ophthalmology & Visual Science, Nanchang 330006, PR China
| | - Haijian Hu
- Affiliated Eye Hospital of Nanchang University, Nanchang University School of Ophthalmology & Optometry, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Research Institute of Ophthalmology & Visual Science, Nanchang 330006, PR China
| | - Ling Zeng
- Affiliated Eye Hospital of Nanchang University, Nanchang University School of Ophthalmology & Optometry, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Research Institute of Ophthalmology & Visual Science, Nanchang 330006, PR China
| | - Hongdou Luo
- Affiliated Eye Hospital of Nanchang University, Nanchang University School of Ophthalmology & Optometry, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Research Institute of Ophthalmology & Visual Science, Nanchang 330006, PR China
| | - Xu Zhang
- Affiliated Eye Hospital of Nanchang University, Nanchang University School of Ophthalmology & Optometry, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Research Institute of Ophthalmology & Visual Science, Nanchang 330006, PR China.
| |
Collapse
|
11
|
Li X, Zhang Z, Wang L, Zhao H, Jia Y, Ma X, Li J, Wang Y, Ma B. Three-phase extraction of polysaccharide from Stropharia rugosoannulata: Process optimization, structural characterization and bioactivities. Front Immunol 2023; 13:994706. [PMID: 36713438 PMCID: PMC9878848 DOI: 10.3389/fimmu.2022.994706] [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: 07/15/2022] [Accepted: 08/23/2022] [Indexed: 01/13/2023] Open
Abstract
The isolation of Stropharia rugosoannulata polysaccharide (SRP) by three-phase extraction was optimized, and its structure and biological activities were identified. The optimal extraction conditions were: mass fraction of ammonium sulfate, 20%; volume ratio of sample solution to t-butanol, 1:1.5; extraction temperature, 35°C. Under these conditions, the yield of SRP was 6.85% ± 0.13%. SRP was found to be composed of glucose (35.79%), galactose (26.80%), glucuronic acid (9.92%), fructose (8.65%), xylose (7.92%), fucose (4.19%), arabinose (3.46%) and rhamnose (3.26%), with the molecular weight of 27.52 kDa. The results of DPPH, hydroxyl, ABTS+ radical scavenging and reducing power tests showed that SRP had good antioxidant capacities. SRP had no cytotoxic effect on RAW264.7 macrophages at the concentrations of 25-200 μg/mL, and could significantly promote phagocytosis activity and cell migration according to CCK-8 assay, phagocytosis assay and cell scratch experiment. SRP can significantly stimulate the transcript expression levels of TNF-α, IL-1β and IL-6, as determined by RT-PCR and Western blot assays. SRP activated the TLR4/NF-κB signaling pathway, and autophagy also occurred. These results suggest that SRP is a safe antioxidant and immunomodulator, and that it can be used in the development of functional foods and/or pharmaceuticals.
Collapse
Affiliation(s)
- Xinxin Li
- Department of Traditional Chinese Medicine, Henan Agricultural University, Zhengzhou, China,School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China,College of Animal medcine, Henan University of Animal husbandry and Economy, Zhengzhou, China
| | - Zhiqiang Zhang
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Li Wang
- Department of Traditional Chinese Medicine, Henan Agricultural University, Zhengzhou, China,*Correspondence: Li Wang, ; Xia Ma, ; Bingji Ma,
| | - Haoqiang Zhao
- Department of Traditional Chinese Medicine, Henan Agricultural University, Zhengzhou, China
| | - Yahui Jia
- Department of Traditional Chinese Medicine, Henan Agricultural University, Zhengzhou, China
| | - Xia Ma
- College of Animal medcine, Henan University of Animal husbandry and Economy, Zhengzhou, China,*Correspondence: Li Wang, ; Xia Ma, ; Bingji Ma,
| | - Jinzhan Li
- Henan Jinlong Mushroom Industry Co. LTD, Shangqiu, China
| | - Yi Wang
- Business Development, GeneGenieDx Corporation, San Jose, CA, United States
| | - Bingji Ma
- Department of Traditional Chinese Medicine, Henan Agricultural University, Zhengzhou, China,*Correspondence: Li Wang, ; Xia Ma, ; Bingji Ma,
| |
Collapse
|
12
|
Lin R, Yu J. The role of NAD + metabolism in macrophages in age-related macular degeneration. Mech Ageing Dev 2023; 209:111755. [PMID: 36435209 DOI: 10.1016/j.mad.2022.111755] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/05/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022]
Abstract
Age-related macular degeneration (AMD) is a leading cause of legal blindness and moderate and severe vision impairment (MSVI) in people older than 50 years. It is classified in various stages including early, intermediate, and late stage. In the early stages, innate immune system, especially macrophages, play an essential part in disease onset and progression. NAD+ is an essential coenzyme involved in cellular senescence and immune cell function, and its role in age-related diseases is gaining increasing attention. The imbalance between the NAD+ synthesis and consumption causes the fluctuation of intracellular NAD+ level which determines the polarization fate of macrophages. In AMD, the over-expression of NAD+-consuming enzymes in macrophages leads to declining of NAD+ concentrations in the microenvironment. This phenomenon triggers the activation of inflammatory pathways in macrophages, positive feedback aggregation of inflammatory cells and accumulation of reactive oxygen species (ROS). This review details the role of NAD+ metabolism in macrophages and molecular mechanisms during AMD. The selected pathways were identified as potential targets for intervention in AMD, pending further investigation.
Collapse
Affiliation(s)
- Ruoyi Lin
- Department of Ophthalmology, the Tenth People's Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai 200072, China
| | - Jing Yu
- Department of Ophthalmology, the Tenth People's Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai 200072, China; Department of Ophthalmology, Bengbu Third People's Hospital, Bengbu, Anhui 233099, China.
| |
Collapse
|
13
|
Qin M, Xie Z, Cao T, Wang Z, Zhang X, Wang F, Wei W, Jin M, Ma J, Zeng L, Wang Y, Pei S, Zhang X. Autophagy in Rat Müller Glial Cells Is Modulated by the Sirtuin 4/AMPK/mTOR Pathway and Induces Apoptosis under Oxidative Stress. Cells 2022; 11:cells11172645. [PMID: 36078054 PMCID: PMC9454555 DOI: 10.3390/cells11172645] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 08/14/2022] [Accepted: 08/18/2022] [Indexed: 11/30/2022] Open
Abstract
Müller glial cells (MGCs) are a group of glial cells in the retina that provide essential support to retinal neurons; however, the understanding of MGC apoptosis and autophagy remains limited. This study was aimed at investigating the role of autophagy in MGCs under normal and oxidative conditions, and identifying the underlying mechanisms. In addition, the sirtuin 4 (SIRT4)-mediated signaling pathway was observed to regulate the autophagic process in MGCs. To assess the effect of autophagy on MGC mitochondrial function and survival, we treated rMC-1 cells—rat-derived Müller glial cells—with rapamycin and 3-methyladenine (3-MA), and found that MGC death was not induced by such treatment, while autophagic dysfunction could increase MGC apoptosis under oxidative stress, as reflected by the expression level of cleaved caspase 3 and PI staining. In addition, the downregulation of autophagy by 3-MA could influence the morphology of the mitochondrial network structure, the mitochondrial membrane potential, and generation of reactive oxygen species (ROS) under oxidative stress. Moreover, SIRT4 depletion enhanced autophagosome formation, as verified by an increase in the LC3 II/I ratio and a decrease in the expression of SQSTM1/p62, and vice versa. The inhibition of AMPK phosphorylation by compound C could reverse these changes in LC3 II/I and SQSTM1/p62 caused by SIRT4 knockdown. Our research concludes that MGCs can endure autophagic dysfunction in the absence of oxidative stress, while the downregulation of autophagy can cause MGCs to become more sensitized to oxidative stress. Simultaneous exposure to oxidative stress and autophagic dysfunction in MGCs can result in a pronounced impairment of cell survival. Mechanically, SIRT4 depletion can activate the autophagic process in MGCs by regulating the AMPK–mTOR signaling pathway.
Collapse
Affiliation(s)
- Mengqi Qin
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center of Ophthalmic Disease, Affiliated Eye Hospital of Nanchang University, Nanchang 330006, China
| | - Zhi Xie
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center of Ophthalmic Disease, Affiliated Eye Hospital of Nanchang University, Nanchang 330006, China
| | - Ting Cao
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center of Ophthalmic Disease, Affiliated Eye Hospital of Nanchang University, Nanchang 330006, China
| | - Zhiruo Wang
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center of Ophthalmic Disease, Affiliated Eye Hospital of Nanchang University, Nanchang 330006, China
| | - Xiaoyu Zhang
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center of Ophthalmic Disease, Affiliated Eye Hospital of Nanchang University, Nanchang 330006, China
| | - Feifei Wang
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center of Ophthalmic Disease, Affiliated Eye Hospital of Nanchang University, Nanchang 330006, China
| | - Wei Wei
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center of Ophthalmic Disease, Affiliated Eye Hospital of Nanchang University, Nanchang 330006, China
| | - Ming Jin
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center of Ophthalmic Disease, Affiliated Eye Hospital of Nanchang University, Nanchang 330006, China
| | - Jingyuan Ma
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center of Ophthalmic Disease, Affiliated Eye Hospital of Nanchang University, Nanchang 330006, China
- Queen Mary School, Nanchang University, Nanchang 330006, China
| | - Ling Zeng
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center of Ophthalmic Disease, Affiliated Eye Hospital of Nanchang University, Nanchang 330006, China
| | - Yanan Wang
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center of Ophthalmic Disease, Affiliated Eye Hospital of Nanchang University, Nanchang 330006, China
| | - Shaonan Pei
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center of Ophthalmic Disease, Affiliated Eye Hospital of Nanchang University, Nanchang 330006, China
| | - Xu Zhang
- Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center of Ophthalmic Disease, Affiliated Eye Hospital of Nanchang University, Nanchang 330006, China
- Correspondence:
| |
Collapse
|
14
|
Ross AG, Chaqour B, McDougald DS, Dine KE, Duong TT, Shindler RE, Yue J, Liu T, Shindler KS. Selective Upregulation of SIRT1 Expression in Retinal Ganglion Cells by AAV-Mediated Gene Delivery Increases Neuronal Cell Survival and Alleviates Axon Demyelination Associated with Optic Neuritis. Biomolecules 2022; 12:830. [PMID: 35740955 PMCID: PMC9221096 DOI: 10.3390/biom12060830] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/07/2022] [Accepted: 06/11/2022] [Indexed: 11/16/2022] Open
Abstract
Optic neuritis (ON), the most common ocular manifestation of multiple sclerosis, is an autoimmune inflammatory demyelinating disease also characterized by degeneration of retinal ganglion cells (RGCs) and their axons, which commonly leads to visual impairment despite attempted treatments. Although ON disease etiology is not known, changes in the redox system and exacerbated optic nerve inflammation play a major role in the pathogenesis of the disease. Silent information regulator 1 (sirtuin-1/SIRT1) is a ubiquitously expressed NAD+-dependent deacetylase, which functions to reduce/prevent both oxidative stress and inflammation in various tissues. Non-specific upregulation of SIRT1 by pharmacologic and genetic approaches attenuates RGC loss in experimental ON. Herein, we hypothesized that targeted expression of SIRT1 selectively in RGCs using an adeno-associated virus (AAV) vector as a delivery vehicle is an effective approach to reducing neurodegeneration and preserving vision in ON. We tested this hypothesis through intravitreal injection of AAV7m8.SNCG.SIRT1, an AAV2-derived vector optimized for highly efficient SIRT1 transgene transfer and protein expression into RGCs in mice with experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis that recapitulates optic neuritis RGC loss and axon demyelination. Our data show that EAE mice injected with a control vehicle exhibit progressive alteration of visual function reflected by decreasing optokinetic response (OKR) scores, whereas comparatively, AAV7m8.SNCG.SIRT1-injected EAE mice maintain higher OKR scores, suggesting that SIRT1 reduces the visual deficit imparted by EAE. Consistent with this, RGC survival determined by immunolabeling is increased and axon demyelination is decreased in the AAV7m8.SNCG.SIRT1 RGC-injected group of EAE mice compared to the mouse EAE counterpart injected with a vehicle or with control vector AAV7m8.SNCG.eGFP. However, immune cell infiltration of the optic nerve is not significantly different among all EAE groups of mice injected with either vehicle or AAV7m8.SNCG.SIRT1. We conclude that despite minimally affecting the inflammatory response in the optic nerve, AAV7m8-mediated SIRT1 transfer into RGCs has a neuroprotective potential against RGC loss, axon demyelination and vison deficits associated with EAE. Together, these data suggest that SIRT1 exerts direct effects on RGC survival and function.
Collapse
Affiliation(s)
- Ahmara G. Ross
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.G.R.); (B.C.); (D.S.M.); (K.E.D.); (T.T.D.); (R.E.S.); (J.Y.); (T.L.)
- F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Brahim Chaqour
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.G.R.); (B.C.); (D.S.M.); (K.E.D.); (T.T.D.); (R.E.S.); (J.Y.); (T.L.)
- F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Devin S. McDougald
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.G.R.); (B.C.); (D.S.M.); (K.E.D.); (T.T.D.); (R.E.S.); (J.Y.); (T.L.)
- F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Kimberly E. Dine
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.G.R.); (B.C.); (D.S.M.); (K.E.D.); (T.T.D.); (R.E.S.); (J.Y.); (T.L.)
- F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Thu T. Duong
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.G.R.); (B.C.); (D.S.M.); (K.E.D.); (T.T.D.); (R.E.S.); (J.Y.); (T.L.)
- F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Ryan E. Shindler
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.G.R.); (B.C.); (D.S.M.); (K.E.D.); (T.T.D.); (R.E.S.); (J.Y.); (T.L.)
- F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jipeng Yue
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.G.R.); (B.C.); (D.S.M.); (K.E.D.); (T.T.D.); (R.E.S.); (J.Y.); (T.L.)
- F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Tehui Liu
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.G.R.); (B.C.); (D.S.M.); (K.E.D.); (T.T.D.); (R.E.S.); (J.Y.); (T.L.)
- F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Kenneth S. Shindler
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.G.R.); (B.C.); (D.S.M.); (K.E.D.); (T.T.D.); (R.E.S.); (J.Y.); (T.L.)
- F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|