1
|
Zhu F, Dan T, Hua S. KEAP1-NRF2/HO-1 Pathway Promotes Ferroptosis and Neuronal Injury in Schizophrenia. Brain Behav 2025; 15:e70311. [PMID: 40021790 PMCID: PMC11870791 DOI: 10.1002/brb3.70311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 12/31/2024] [Accepted: 01/16/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND This study investigates the role of the KEAP1-NRF2/HO-1 signaling pathway in inducing ferroptosis and contributing to neuronal damage in schizophrenia. METHODS We retrieved schizophrenia-related data and ferroptosis-related genes from the RNA microarray dataset GSE27383 and FerrDB database, respectively. Bioinformatics data identified KEAP1 as a downregulated gene, which was validated using qRT-PCR and Western blot. We assessed intracellular Fe2⁺ content, MDA levels, GSH, and GPX4 in the prefrontal cortex and peripheral blood mononuclear cells (PBMCs) of patients with schizophrenia. Cortical interneurons (cINs) were generated from human-induced pluripotent stem cells (hiPSCs) of patients with schizophrenia and used to explore KEAP1 alterations during neurodevelopment. In addition, KEAP1 overexpression was induced in cINs via transfection with pcDNA KEAP1. The intracellular Fe⁺ levels, oxidative stress indicators, lipid peroxidation, and inflammatory cytokines were measured after transfection. To investigate molecular mechanisms, KI696-a high-affinity probe that disrupts the KEAP1-NRF2 interaction-was applied, and changes in oxidative stress, lipid peroxidation (C11-BODIPY staining), iron metabolism, and inflammatory pathways were evaluated. RESULTS Patients with schizophrenia exhibited underexpression of KEAP1, a key regulator of ferroptosis, along with elevated intracellular Fe2⁺ levels and increased MDA concentrations, indicating enhanced lipid peroxidation and oxidative stress. Reduced GPX4 activity and GSH levels were also observed, suggesting an increased susceptibility to ferroptosis. To further explore this, cINs derived from hiPSCs of patients with schizophrenia were studied. These cells showed decreased KEAP1 expression. Overexpression of KEAP1 in cINs led to a reduction in intracellular Fe2⁺ concentrations and oxidative damage, highlighting KEAP1's regulatory role in ferroptosis. In addition, treatment with KI696 induced significant alterations in pathways related to oxidative stress, iron metabolism, antioxidant defenses, and inflammation. CONCLUSION Our findings indicate that the KEAP1-NRF2/HO-1 pathway contributes to ferroptosis and neuronal injury in schizophrenia.
Collapse
Affiliation(s)
- Feng Zhu
- Department of PsychiatryThe Second Affiliated Hospital of Hubei University of Science and TechnologyXianningHubeiChina
| | - Tangqun Dan
- Department of PsychiatryThe Second Affiliated Hospital of Hubei University of Science and TechnologyXianningHubeiChina
| | - Shuguang Hua
- Department of PsychiatryThe Second Affiliated Hospital of Hubei University of Science and TechnologyXianningHubeiChina
| |
Collapse
|
2
|
Jiang H, Liu Y, Yu Y, Yan Y. Sirtuin 1 Suppresses Hydrogen Peroxide-Induced Senescence and Promotes Viability and Migration in Lens Epithelial Cells by Inhibiting Forkhead Box Protein O1/Toll-Like Receptor 4 Pathway. J Biochem Mol Toxicol 2025; 39:e70150. [PMID: 39866090 DOI: 10.1002/jbt.70150] [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: 10/09/2024] [Revised: 12/24/2024] [Accepted: 01/16/2025] [Indexed: 01/28/2025]
Abstract
Age-related cataracts (ARCs) are associated with increased oxidative stress and cellular senescence. Our objective is to investigate the function of Sirtuin 1 (SIRT1) within ARCs. In ARCs tissues and H2O2-treated lens epithelial cells (LECs), the expression levels of SIRT1 were examined. Senescence-associated β-galactosidase (SA-β-gal) staining was employed to evaluate cellular senescence. The Cell Counting Kit-8 assay was employed to measure viability. A wound healing assay was performed to assess migratory capacity in LECs. Oxidative stress-related indicators were determined by enzyme-linked immunosorbent assay kits. Additionally, the Coxpresdb and GeneCards databases were utilized to identify downstream pathways of SIRT1 in ARCs. The expression levels of protein and mRNA were detected using western blot and real-time quantitative polymerase chain reaction, respectively. The expression of SIRT1 was downregulated in ARCs tissues with an increase in reactive oxygen species. In H2O2-induced LECs, SIRT1 was downregulated and its overexpression inhibited oxidative stress and cellular senescence while promoting viability and migration. Furthermore, FoxO1/TLR4 pathway was screened out as the key pathway of SIRT1, which was activated in H2O2-induced LECs senescence. Overexpression of SIRT1 suppressed FoxO1/TLR4 pathway. Further research demonstrated that the activation of FoxO1/TLR4 pathway reversed the inhibitory role of SIRT1 in oxidative stress-induced cellular senescence and the promotion effect of SIRT1 on viability and migration in H2O2-induced LECs. SIRT1 inhibits oxidative stress-induced cellular senescence and promotes the viability and migration in H2O2-induced LECs via suppressing FoxO1/TLR4 pathway.
Collapse
Affiliation(s)
- Hongda Jiang
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yuting Liu
- Department of Ophthalmology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yinggui Yu
- Department of Ophthalmology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yu Yan
- Department of Ophthalmology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
3
|
Fu L, Yang Q, Han Y, Sun F, Jin J, Wang J. Slit2 Promotes H 2O 2-Induced Lens Epithelial Cells Oxidative Damage and Age-Related Cataract. Curr Eye Res 2025; 50:41-50. [PMID: 39143744 DOI: 10.1080/02713683.2024.2388698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 05/14/2024] [Accepted: 08/01/2024] [Indexed: 08/16/2024]
Abstract
PURPOSE To analyze the role of Slit2 in lens epithelial cell oxidative damage and its underlying mechanism. METHODS Human lens epithelial cells (SRA01/04 cells) and rat transparent lens were cultured with H2O2 to establish cell oxidative stress models and rat cataract models. Immunohistochemistry, quantitative real-time polymerase chain reaction (qRT-PCR), and Western blot assays were employed to detect Slit2 levels within age-related cataracts(ARC) lens anterior capsule samples, rat cataract models, and cell oxidative stress models. In this study, qRT-PCR and Western blot assays were performed to derermine E-cadherin, N-cadherin, occludens1(ZO-1), α-SMA(α‑smooth muscle actin), Bcl-2, Bax, p-AKT, and AKT levels. In addition, Flow cytometry were performed to examine reactive oxygen species (ROS) and cell apoptosis. Cell viability, invasion, and migration were detected by CCK8, Transwell, and Wound healing. RESULTS Increased expression of Slit2 was found in ARC lens anterior capsule samples, H2O2-induced rat cataract models, and Human lens epithelial cells (HLECs) oxidative stress models. H2O2 significantly increased cell apoptosis and ROS generation, also accelerating cell migration, invasion, and epithelial-mesenchymal transition (EMT). In addition, H2O2 treatment repressed AKT phosphorylation and cell viability. Knock-down of Slit2 promoted cell viability and AKT phosphorylation levels, as well as repressed cell invasion, migration, apoptosis, ROS production and EMT. CONCLUSION Slit2 promoted lens epithelial cells oxidative stress damage via the AKT signalling pathways, providing a novel insight in ARC treatment.
Collapse
Affiliation(s)
- Lingzhi Fu
- Department of Ophthalmology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical University, Bengbu, Anhui, China
| | - Qing Yang
- Department of Ophthalmology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical University, Bengbu, Anhui, China
| | - Yuanyuan Han
- Department of Ophthalmology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Feng Sun
- Department of Ophthalmology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Jiacheng Jin
- School of Life Sciences, Bengbu Medical University, Bengbu, Anhui, China
| | - Jianfeng Wang
- Department of Ophthalmology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| |
Collapse
|
4
|
Nath KA, Juncos LA, Singh RD, Grande JP, Croatt AJ, Ackerman AW, Kanamori KS, Adams CM, Tchkonia T, Kirkland JL, Katusic ZS. The Occurrence of Senescence in the Arteriovenous Fistula in the Rat. KIDNEY360 2025; 6:27-37. [PMID: 39418108 PMCID: PMC11793188 DOI: 10.34067/kid.0000000605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024]
Abstract
Key Points The rat arteriovenous fistula (AVF) model exhibits marked upregulation of p16Ink4a and p21Cip1 and multiple markers of senescence. Fisetin, an established vasoprotective senolytic agent, when administered for 3 weeks, increases AVF blood flow and AVF outward remodeling. Heme is shown to be a novel prosenescence metabolite, and when chronically administered, it decreases AVF blood flow. Background Maturational failure of dialysis arteriovenous fistulas (AVFs) not uncommonly occurs and is of considerable and timely importance. Our prior studies demonstrate that senescence, a phenotypic process that promotes vascular and other diseases, occurs in the murine AVF. In this study, we examined whether senescence also occurs in the rat AVF model and the effect of compounds that inhibit or accelerate senescence. Methods The rat AVF was created in the femoral vessels by an end vein-side artery anastomosis. In the AVF, we assessed the expression of critical drivers of senescence, specifically, the cell cycle inhibitors p16Ink4a and p21Cip1, and such indices of a senescence phenotype as senescence-associated β -galactosidase (SA-β -gal) activity, SA-β -gal staining, and a senescence-associated secretory phenotype. We examined the effects of compounds that retard or accelerate senescence on AVF blood flow. Results The AVF evinced upregulation of p16Ink4a and p21Cip1 when assessed 3 days after AVF creation. The AVF also demonstrated increased SA-β -gal activity in the artery and vein; staining for SA-β -gal in the AVF artery, anastomosis, and vein; and a prominent senescence-associated secretory phenotype. Fisetin, an established senolytic that is protective in other models of vascular injury, when administered for 3 weeks, increased AVF blood flow and outward remodeling. Hemin, when administered for 3 weeks, decreased AVF blood flow. We demonstrate that hemin is a novel inducer of a senescence phenotype in endothelial cells, as reflected by several senescence indices. However, when administered relatively acutely (for 5 days), hemin increased AVF blood flow by heme oxygenase–dependent mechanisms because the latter was entirely prevented by a competitive inhibitor of heme oxygenase activity. Conclusions The rat AVF exhibits senescence within 3 days of its creation. Chronic administration of a senolytic compound (fisetin) increases AVF blood flow, whereas chronic administration of a prosenescence compound (hemin) decreases AVF blood flow.
Collapse
Affiliation(s)
- Karl A. Nath
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Luis A. Juncos
- University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Raman Deep Singh
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Joseph P. Grande
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Anthony J. Croatt
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Allan W. Ackerman
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Karina S. Kanamori
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Christopher M. Adams
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Tamara Tchkonia
- Division of Endocrinology, Department of Medicine, Center for Advanced Gerotherapeutics, Cedars-Sinai Health Sciences Center, Los Angeles, California
| | - James L. Kirkland
- Division of Endocrinology, Department of Medicine, Center for Advanced Gerotherapeutics, Cedars-Sinai Health Sciences Center, Los Angeles, California
| | - Zvonimir S. Katusic
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
5
|
Dong S, Zhang J, Fu Y, Tang G, Chen J, Sun D, Qi Y, Zhou N. METTL3-mediated m6A modification of SIRT1 mRNA affects the progression of diabetic cataracts through cellular autophagy and senescence. J Transl Med 2024; 22:865. [PMID: 39334185 PMCID: PMC11429169 DOI: 10.1186/s12967-024-05691-w] [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: 07/23/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND The increasing incidence of diabetes mellitus has established diabetic cataracts (DC) as a significant worldwide public health issue. The mechanisms underlying DC remain unknown, and effective prevention and treatment strategies are lacking. Accordingly, we aimed to explore the role and mechanism behind N6-methyladenosine (m6A) in DC progression. METHODS Methyltransferase-like 3 (METTL3), p21, Beclin1, LC3, and p62 expression levels were measured in human tissues. This study assessed total m6A levels and common m6A-regulated biomarkers in both in vitro and in vivo DC models. Autophagy flux was detected in vitro through Ad-mCherry-GFP-LC3B and Monodansylcadaverine (MDC) staining. Cellular senescence was assessed utilizing the senescence-associated β-galactosidase (SA-β-Gal) assay. Furthermore, the effect of METTL3 on SIRT1 mRNA modification was demonstrated, and its mechanism was elucidated using RT-qPCR, western blot, RNA stability assays, and RIP analysis. RESULTS METTL3, p21, and p62 expression levels were elevated in lens epithelial cells (LECs) from DC patients, while Beclin1 and LC3 levels were reduced. Silencing METTL3-mediated m6A modifications restored high-glucose-induced autophagy inhibition and prevented premature senescence in LECs. Notably, SIRT1720 and Metformin significantly enhanced autophagosome generation and delayed cellular senescence. The m6A-reading protein YTHDF2 bound to m6A modifications, and YTHDF2 silencing significantly reduced METTL3-mediated SIRT1 inactivation. CONCLUSIONS METTL3 induces senescence in DC by destabilizing SIRT1 mRNA in an m6A-YTHDF2-dependent manner. The METTL3-YTHDF2-SIRT1 axis is a key target and potential pathogenic mechanism in DC.
Collapse
Affiliation(s)
- Su Dong
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Jiajia Zhang
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Yushan Fu
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Gege Tang
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Jianfeng Chen
- Laboratory Animal Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Dawei Sun
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China.
| | - Yanhua Qi
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China.
| | - Nan Zhou
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China.
| |
Collapse
|
6
|
Jiang Y, Xiao X, Sun W, Wang Y, Li S, Jia X, Wang P, Hejtmancik JF, Zhang Q. Clinical and genetic risk factors underlying severe consequence identified in 75 families with unilateral high myopia. J Transl Med 2024; 22:75. [PMID: 38243264 PMCID: PMC10797748 DOI: 10.1186/s12967-024-04886-5] [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: 09/14/2023] [Accepted: 01/11/2024] [Indexed: 01/21/2024] Open
Abstract
BACKGROUNDS Unilateral high myopia (uHM), commonly observed in patients with retinal diseases or only with high myopia, is frequently associated with amblyopia with poor prognosis. This study aims to reveal the clinical and genetic spectrum of uHM in a large Chinese cohort. METHODS A total of 75 probands with simplex uHM were included in our Pediatric and Genetic Eye Clinic. Patients with significant posterior anomalies other than myopic fundus changes were excluded. Variants were detected by exome sequencing and then analyzed through multiple-step bioinformatic and co-segregation analysis and finally confirmed by Sanger sequencing. Genetic findings were correlated with associated clinical data for analysis. RESULTS Among the 75 probands with a mean age of 6.21 ± 4.70 years at the presentation, myopic fundus of C1 and C2 was observed in 73 (97.3%) probands. Surprisingly, specific peripheral changes were identified in 63 eyes involving 36 (48.0%) probands after extensive examination, including peripheral retinal avascular zone (74.6%, 47/63 eyes), neovascularization (54.0%), fluorescein leakage (31.7%), peripheral pigmentary changes (31.7%), and others. Exome sequencing identified 21 potential pathogenic variants of 13 genes in 20 of 75 (26.7%) probands, including genes for Stickler syndrome (COL11A1 and COL2A1; 6/20), FEVR (FZD4, LRP5, and TSPAN12; 5/20), and others (FBN1, GPR179, ZEB2, PAX6, GPR143, OPN1LW, FRMD7, and CACNA1F; 9/20). For the peripheral retinal changes in the 20 probands, variants in Stickler syndrome-related genes were predominantly associated with retinal pigmentary changes, lattice degeneration, and retinal avascular region, while variants in genes related to FEVR were mainly associated with the avascular zone, neovascularization, and fluorescein leakage. CONCLUSIONS Genetic defects were identified in about one-fourth of simplex uHM patients in which significant consequences may be hidden under a classic myopic fundus in up to half. To our knowledge, this is the first systematic genetic study on simplex uHM to date. In addition to routine care of strabismus and amblyopia, careful examination of the peripheral retina and genetic screening is warranted for patients with uHM in order to identify signs of risk for retinal detachment and other complications and provide meaningful genetic counseling.
Collapse
Affiliation(s)
- Yi Jiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 54 Xianlie Road, Guangzhou, 510060, China
| | - Xueshan Xiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 54 Xianlie Road, Guangzhou, 510060, China
| | - Wenmin Sun
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 54 Xianlie Road, Guangzhou, 510060, China
| | - Yingwei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 54 Xianlie Road, Guangzhou, 510060, China
| | - Shiqiang Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 54 Xianlie Road, Guangzhou, 510060, China
| | - Xiaoyun Jia
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 54 Xianlie Road, Guangzhou, 510060, China
| | - Panfeng Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 54 Xianlie Road, Guangzhou, 510060, China
| | - J Fielding Hejtmancik
- Ophthalmic Molecular Genetics Section, Ophthalmic Genetics and Visual Function Branch, National Eye Institute, Rockville, MD, 20852, USA
| | - Qingjiong Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 54 Xianlie Road, Guangzhou, 510060, China.
| |
Collapse
|