1
|
Luo X, Xue D. Potential mechanisms of epigenetic regulation in diabetic retinopathy from the perspectives of multi-omics. Diabetol Metab Syndr 2025; 17:155. [PMID: 40369608 PMCID: PMC12076923 DOI: 10.1186/s13098-025-01723-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 05/02/2025] [Indexed: 05/16/2025] Open
Abstract
PURPOSE Diabetic retinopathy (DR) is a significant complication of diabetes, with complex pathogenesis involving epigenetic modifications. This study aimed to explore the epigenetic regulatory mechanisms contributing to DR. METHODS DR-related data, including DNA methylation, mRNA, and miRNA expression datasets, were obtained from the Gene Expression Omnibus database. Differential gene expression analysis was performed to identify differentially methylated genes and expressed mRNAs and miRNAs. Cross-analysis established the methylation-expression and miRNA-mRNA regulatory networks. A comprehensive DR-related epigenetic regulatory network was constructed, identifying hub genes. The expression characteristics of these hub genes in various immune cells were examined using single-cell RNA sequencing. RESULTS We identified 10,716 differentially methylated genes, 1,181 differentially expressed mRNAs, and 615 differentially expressed miRNAs in DR. The methylation-expression regulatory network was associated with pathways such as TGF-beta and ErbB signaling. The miRNA regulatory network was linked to pathways related to cellular senescence, adherents junctions, and endocytosis. Five hub genes were identified: TFRC, AP2M1, AP2A1, DAB2, and PPP1CB. Single-cell RNA sequencing revealed specific expression of these genes in particular immune cells, highlighting their potential roles in DR pathogenesis. CONCLUSION This study constructed a comprehensive epigenetic regulatory network for DR and identified key regulatory genes, offering new insights into the molecular mechanisms underlying DR and potential therapeutic targets for diagnosis and treatment.
Collapse
Affiliation(s)
- Xin Luo
- Department of Ophthalmic, Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital Northwest University, Xi'an, 710004, China
| | - Daxi Xue
- Department of Ophthalmic, Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated People's Hospital Northwest University, Xi'an, 710004, China.
| |
Collapse
|
2
|
Jia L, Xiao H, Hao Z, Sun S, Zhao W, Gong Z, Gu W, Wen Y. Senolytic elimination of senescent cells improved periodontal ligament stem cell-based bone regeneration partially through inhibiting YAP. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119921. [PMID: 39971252 DOI: 10.1016/j.bbamcr.2025.119921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 01/28/2025] [Accepted: 02/13/2025] [Indexed: 02/21/2025]
Abstract
Periodontal ligament stem cell (PDLSC)-based tissue engineering is an important method to promote periodontal tissue regeneration. However, PDLSCs are susceptible to the effects of replicative senescence, leading to reduced proliferation and differentiation abilities and weakened tissue regeneration potential. Senolytics (the combination of dasatinib and quercetin) are drugs that inhibit cellular aging through inducing the apoptosis of senescent cells, but whether they have positive effects during the senescence of PDLSCs is unknown. The present study established a long-term in vitro culture model of PDLSCs and then analyzed the effects of senolytics on the senescence, apoptosis, and osteogenic differentiation of PDLSCs in vitro and PDLSC-based tissue regeneration in vivo. The results showed that senolytics delayed the process of aging in prolonged-cultured PDLSCs and promoted the elimination and apoptosis of senescent cells. Moreover, senolytics improved the osteogenic differentiation ability of both young and senescent PDLSCs in vitro and promoted PDLSC-based alveolar bone regeneration in vivo. Furthermore, senolytics inhibited the expression of YAP in senescent PDLSCs. Their antiaging effects were enhanced when combined with the YAP inhibitor verteporfin, but were inhibited when combined with the YAP activator NIBR-LTSi. Taken together, these findings suggest that senolytics promoted the elimination of senescent PDLSCs and enhanced senescent PDLSC-based bone regeneration, partially through the inhibition of YAP expression.
Collapse
Affiliation(s)
- Linglu Jia
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China; Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Han Xiao
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China; Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Zhenghao Hao
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China; Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Shaoqing Sun
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China; Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Wenxi Zhao
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China; Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Zikai Gong
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China; Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Weiting Gu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, China.
| | - Yong Wen
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China; Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China.
| |
Collapse
|
3
|
Shirian JD, Shukla P, Singh RP. Exploring new horizons in neovascular age-related macular degeneration: novel mechanisms of action and future therapeutic avenues. Eye (Lond) 2025; 39:40-44. [PMID: 39379521 PMCID: PMC11733175 DOI: 10.1038/s41433-024-03373-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 08/28/2024] [Accepted: 09/20/2024] [Indexed: 10/10/2024] Open
Abstract
Neovascular age-related macular degeneration (nAMD) can lead to significant vision impairment through the growth of abnormal neovascular membranes in the choroid. Despite advancements with current anti-vascular endothelial growth factor (VEGF) therapies, challenges such as frequent injections, inadequate response, and patient-related concerns persist. Emerging therapeutics aim to reduce vision-loss through a variety of mechanisms. Gene therapies, including RGX-314 and Ixo-vec, express an anti-VEGF protein, and 4D-150, expresses an anti-VEGF protein and a VEGF-C inhibitory miRNA. Anti-VEGF associated therapeutics include OPT-302, targeting VEGF-C and VEGF-D, BI 836880, which inhibits VEGF-A and Ang-2 activity, and Tarcocimab tedromer, inhibiting all VEGF-A isoforms. Agents with novel mechanisms of action include UBX1325, which inhibits an anti-apoptotic protein, Restoret (EYE103), a Wnt agonist, and the tyrosine kinase inhibitors, EYP-1901, OTX-TKI, CLS-AX, and KHK4951.
Collapse
Affiliation(s)
- Jonathan D Shirian
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Center for Ophthalmic Bioinformatics, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Priya Shukla
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Center for Ophthalmic Bioinformatics, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Rishi P Singh
- Center for Ophthalmic Bioinformatics, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA.
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA.
- Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, USA.
- Cleveland Clinic Martin Health, Cleveland Clinic Florida, Stuart, FL, USA.
| |
Collapse
|
4
|
Bi J, Zeng J, Liu X, Mo C, Yao M, Zhang J, Yuan P, Jia B, Xu S. Drug delivery for age-related bone diseases: From therapeutic targets to common and emerging therapeutic strategies. Saudi Pharm J 2024; 32:102209. [PMID: 39697472 PMCID: PMC11653637 DOI: 10.1016/j.jsps.2024.102209] [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: 09/12/2024] [Accepted: 11/22/2024] [Indexed: 12/20/2024] Open
Abstract
With the accumulation of knowledge on aging, people have gradually realized that among the many factors that cause individual aging, the accumulation of aging cells is an essential cause of organ degeneration and, ultimately, age-related diseases. Most cells present in the bone microenvironment gradually age over time, leading to an imbalance of osteogenesis, osteoclastogenesis, adipogenesis, and chondrogenesis. This imbalance contributes to age-related bone loss and the development of age-related bone diseases, such as osteoporosis. Bone aging can prolong the lifespan and delay the development of age-related diseases. Nanoparticles have controllable and stable physical and chemical properties and can precisely target different tissues and organs. By preparing multiple easily modified and biocompatible nanoparticles as different drug delivery carriers, specifically targeting various diseased tissues for controlled-release and sustained-release administration, the delivery efficiency of drugs can be significantly improved, and the toxicity and side effects of drugs can be substantially reduced, thereby improving the therapeutic effect of age-related bone diseases. In addition, other novel anti-aging strategies (such as stem cell exosomes) also have significant scientific and practical significance in anti-aging research on age-related bone diseases. This article reviews the research progress of various nano-drug-loaded particles and emerging anti-aging methods for treating age-related bone diseases, offering new insights and directions for precise targeted clinical therapies.
Collapse
Affiliation(s)
- Jiaming Bi
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiawei Zeng
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaohao Liu
- Department of Periodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Chuzi Mo
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Mingyan Yao
- Department of Endocrinology, Baoding No.1 Central Hospital, Baoding, China
| | - Jing Zhang
- Department of Cardiology, Affiliated Hospital of Hebei University, Baoding, China
| | - Peiyan Yuan
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Bo Jia
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Shuaimei Xu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
5
|
Riessland M, Ximerakis M, Jarjour AA, Zhang B, Orr ME. Therapeutic targeting of senescent cells in the CNS. Nat Rev Drug Discov 2024; 23:817-837. [PMID: 39349637 PMCID: PMC11927922 DOI: 10.1038/s41573-024-01033-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2024] [Indexed: 11/01/2024]
Abstract
Senescent cells accumulate throughout the body with advanced age, diseases and chronic conditions. They negatively impact health and function of multiple systems, including the central nervous system (CNS). Therapies that target senescent cells, broadly referred to as senotherapeutics, recently emerged as potentially important treatment strategies for the CNS. Promising therapeutic approaches involve clearing senescent cells by disarming their pro-survival pathways with 'senolytics'; or dampening their toxic senescence-associated secretory phenotype (SASP) using 'senomorphics'. Following the pioneering discovery of first-generation senolytics dasatinib and quercetin, dozens of additional therapies have been identified, and several promising targets are under investigation. Although potentially transformative, senotherapies are still in early stages and require thorough testing to ensure reliable target engagement, specificity, safety and efficacy. The limited brain penetrance and potential toxic side effects of CNS-acting senotherapeutics pose challenges for drug development and translation to the clinic. This Review assesses the potential impact of senotherapeutics for neurological conditions by summarizing preclinical evidence, innovative methods for target and biomarker identification, academic and industry drug development pipelines and progress in clinical trials.
Collapse
Affiliation(s)
- Markus Riessland
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, USA
- Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY, USA
| | | | | | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miranda E Orr
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
- Salisbury VA Medical Center, Salisbury, NC, USA.
| |
Collapse
|
6
|
Sakini ASA, Hamid AK, Alkhuzaie ZA, Al-Aish ST, Al-Zubaidi S, Tayem AA, Alobi MA, Sakini ASA, Al-Aish RT, Al-Shami K, Hanifa H, Khunda SS. Diabetic macular edema (DME): dissecting pathogenesis, prognostication, diagnostic modalities along with current and futuristic therapeutic insights. Int J Retina Vitreous 2024; 10:83. [PMID: 39468614 PMCID: PMC11514910 DOI: 10.1186/s40942-024-00603-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 10/17/2024] [Indexed: 10/30/2024] Open
Abstract
One of the most common health concerns disturbing people within working years globally is diabetes mellitus (DM). One well-known consequence of DM is vascular damage, which can manifest as macro- and microangiopathy affecting the ocular retina. Therefore, Diabetic macular edema (DME) is a major sight-threatening complication of diabetic retinopathy (DR) worldwide. It is the most prevalent cause of significant vision impairment in diabetic patients. Long-term vision loss can be avoided by following early DME treatment guidelines in everyday life. Hence, there are various therapeutic approaches for DME management. Currently, the first-line treatment for DME is anti-VEGF family drugs, such as ranibizumab, brolucizumab, bevacizumab, and aflibercept. Nevertheless, relapses of the disease, inadequate response, and resistance during anti-VEGF therapy are still seen because of the intricate pathophysiological foundation of the disease. Consequently, there is an excellent requirement for therapeutic approaches to advance and become better at controlling diseases more satisfactorily and require fewer treatments overall. We conducted a thorough literature search in the current review to present a comprehensive overview of the primary data about the current DME therapeutic agents. We also covered the novel advances in DME management and probable future treatments being investigated and developed. This review recommended that Large clinical trials should afford sufficient evidence to support these innovative treatment modalities.
Collapse
Affiliation(s)
| | | | - Zainab A Alkhuzaie
- Clinical Teaching Fellow, College of Medicine, University of Kufa, Al-Najaf, Iraq
| | - Sandra Thair Al-Aish
- Department of Surgery, College of Medicine, University of Baghdad, Baghdad, Iraq
| | - Shahad Al-Zubaidi
- Clinical Teaching Fellow, University of Baghdad, Al-Kindy Medical College, Baghdad, Iraq
| | | | | | | | - Rami Thair Al-Aish
- Department of Surgery, College of Medicine, University of Baghdad, Baghdad, Iraq
| | - Khayry Al-Shami
- Department of Clinical Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid, Jordan
| | - Hamdah Hanifa
- Faculty of Medicine, University of Kalamoon, Al-Nabk, Syria.
| | - Sara S Khunda
- Department of Internal Medicine, Baghdad Medical City, Baghdad, Iraq
| |
Collapse
|
7
|
Zhang X, Zhang F, Xu X. Single-cell RNA sequencing in exploring the pathogenesis of diabetic retinopathy. Clin Transl Med 2024; 14:e1751. [PMID: 38946005 PMCID: PMC11214886 DOI: 10.1002/ctm2.1751] [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: 03/13/2024] [Revised: 06/10/2024] [Accepted: 06/16/2024] [Indexed: 07/02/2024] Open
Abstract
Diabetic retinopathy (DR) is a leading cause of irreversible blindness in the working-age populations. Despite decades of research on the pathogenesis of DR for clinical care, a comprehensive understanding of the condition is still lacking due to the intricate cellular diversity and molecular heterogeneity involved. Single-cell RNA sequencing (scRNA-seq) has made the high-throughput molecular profiling of cells across modalities possible which has provided valuable insights into complex biological systems. In this review, we summarise the application of scRNA-seq in investigating the pathogenesis of DR, focusing on four aspects. These include the identification of differentially expressed genes, characterisation of key cell subpopulations and reconstruction of developmental 'trajectories' to unveil their state transition, exploration of complex cell‒cell communication in DR and integration of scRNA-seq with genome-wide association studies to identify cell types that are most closely related to DR risk genetic loci. Finally, we discuss the future challenges and expectations associated with studying DR using scRNA-seq. We anticipate that scRNA-seq will facilitate the discovery of mechanisms and new treatment targets in the clinical care landscape for patients with DR. KEY POINTS: Progress in scRNA-seq for diabetic retinopathy (DR) research includes studies on DR patients, non-human primates, and the prevalent mouse models. scRNA-seq facilitates the identification of differentially expressed genes, pivotal cell subpopulations, and complex cell-cell interactions in DR at single-cell level. Future scRNA-seq applications in DR should target specific patient subsets and integrate with single-cell and spatial multi-omics approaches.
Collapse
Affiliation(s)
- Xinzi Zhang
- National Clinical Research Center for Eye DiseasesDepartment of OphthalmologyShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Eye Institute of Shanghai Jiao Tong University SchoolShanghaiChina
- Shanghai Key Laboratory of Ocular Fundus DiseasesShanghaiChina
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye DiseasesShanghaiChina
| | - Fang Zhang
- National Clinical Research Center for Eye DiseasesDepartment of OphthalmologyShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Eye Institute of Shanghai Jiao Tong University SchoolShanghaiChina
- Shanghai Key Laboratory of Ocular Fundus DiseasesShanghaiChina
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye DiseasesShanghaiChina
| | - Xun Xu
- National Clinical Research Center for Eye DiseasesDepartment of OphthalmologyShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Eye Institute of Shanghai Jiao Tong University SchoolShanghaiChina
- Shanghai Key Laboratory of Ocular Fundus DiseasesShanghaiChina
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye DiseasesShanghaiChina
| |
Collapse
|
8
|
Zhang G, Samarawickrama PN, Gui L, Ma Y, Cao M, Zhu H, Li W, Yang H, Li K, Yang Y, Zhu E, Li W, He Y. Revolutionizing Diabetic Foot Ulcer Care: The Senotherapeutic Approach. Aging Dis 2024; 16:946-970. [PMID: 38739931 PMCID: PMC11964433 DOI: 10.14336/ad.2024.0065] [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: 01/17/2024] [Accepted: 04/16/2024] [Indexed: 05/16/2024] Open
Abstract
Diabetic foot ulcers (DFUs) are a prevalent and profoundly debilitating complication that afflicts individuals with diabetes mellitus (DM). These ulcers are associated with substantial morbidity, recurrence rates, disability, and mortality, imposing substantial economic, psychological, and medical burdens. Timely detection and intervention can mitigate the morbidity and disparities linked to DFU. Nevertheless, current therapeutic approaches for DFU continue to grapple with multifaceted limitations. A growing body of evidence emphasizes the crucial role of cellular senescence in the pathogenesis of chronic wounds. Interventions that try to delay cellular senescence, eliminate senescent cells (SnCs), or suppress the senescence-associated secretory phenotype (SASP) have shown promise for helping chronic wounds to heal. In this context, targeting cellular senescence emerges as a novel therapeutic strategy for DFU. In this comprehensive review, we look at the pathology and treatment of DFU in a systematic way. We also explain the growing importance of investigating SnCs in DFU and highlight the great potential of senotherapeutics that target SnCs in DFU treatment. The development of efficacious and safe senotherapeutics represents a pioneering therapeutic approach aimed at enhancing the quality of life for individuals affected by DFU.
Collapse
Affiliation(s)
- Guiqin Zhang
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China.
| | - Priyadarshani Nadeeshika Samarawickrama
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China.
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China.
| | - Li Gui
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China.
| | - Yuan Ma
- Department of Orthopedics, the Third People’s Hospital of Yunnan Province, Kunming, Yunnan 650011, China.
| | - Mei Cao
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China.
| | - Hong Zhu
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China.
| | - Wei Li
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China.
| | - Honglin Yang
- Department of Orthopedics, the Third People’s Hospital of Yunnan Province, Kunming, Yunnan 650011, China.
| | - Kecheng Li
- Department of Orthopedics, the Third People’s Hospital of Yunnan Province, Kunming, Yunnan 650011, China.
| | - Yang Yang
- Department of Biochemistry & Structural Biology, University of Texas Health Science Center, San Antonio, TX 78229, USA.
| | - Enfang Zhu
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China.
| | - Wen Li
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China.
| | - Yonghan He
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China.
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China.
| |
Collapse
|
9
|
Petr MA, Matiyevskaya F, Osborne B, Berglind M, Reves S, Zhang B, Ezra MB, Carmona-Marin LM, Syadzha MF, Mediavilla MC, Keijzers G, Bakula D, Mkrtchyan GV, Scheibye-Knudsen M. Pharmacological interventions in human aging. Ageing Res Rev 2024; 95:102213. [PMID: 38309591 DOI: 10.1016/j.arr.2024.102213] [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: 08/09/2023] [Revised: 01/15/2024] [Accepted: 01/30/2024] [Indexed: 02/05/2024]
Abstract
Pharmacological interventions are emerging as potential avenues of alleviating age-related disease. However, the knowledge of ongoing clinical trials as they relate to aging and pharmacological interventions is dispersed across a variety of mediums. In this review we summarize 136 age-related clinical trials that have been completed or are ongoing. Furthermore, we establish a database that describe the trials (AgingDB, www.agingdb.com) keeping track of the previous and ongoing clinical trials, alongside their outcomes. The aim of this review and database is to give people the ability to easily query for their trial of interest and stay up to date on the latest results. In sum, herein we give an overview of the current pharmacological strategies that have been applied to target human aging.
Collapse
Affiliation(s)
- Michael Angelo Petr
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Frida Matiyevskaya
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Brenna Osborne
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Magnus Berglind
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Simon Reves
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Bin Zhang
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Michael Ben Ezra
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Lina Maria Carmona-Marin
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Muhammad Farraz Syadzha
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Marta Cortés Mediavilla
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Guido Keijzers
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Daniela Bakula
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Garik V Mkrtchyan
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Morten Scheibye-Knudsen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark.
| |
Collapse
|
10
|
Fraile-Martinez O, De Leon-Oliva D, Boaru DL, De Castro-Martinez P, Garcia-Montero C, Barrena-Blázquez S, García-García J, García-Honduvilla N, Alvarez-Mon M, Lopez-Gonzalez L, Diaz-Pedrero R, Guijarro LG, Ortega MA. Connecting epigenetics and inflammation in vascular senescence: state of the art, biomarkers and senotherapeutics. Front Genet 2024; 15:1345459. [PMID: 38469117 PMCID: PMC10925776 DOI: 10.3389/fgene.2024.1345459] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/15/2024] [Indexed: 03/13/2024] Open
Abstract
Vascular diseases pose major health challenges, and understanding their underlying molecular mechanisms is essential to advance therapeutic interventions. Cellular senescence, a hallmark of aging, is a cellular state characterized by cell-cycle arrest, a senescence-associated secretory phenotype macromolecular damage, and metabolic dysregulation. Vascular senescence has been demonstrated to play a key role in different vascular diseases, such as atherosclerosis, peripheral arterial disease, hypertension, stroke, diabetes, chronic venous disease, and venous ulcers. Even though cellular senescence was first described in 1961, significant gaps persist in comprehending the epigenetic mechanisms driving vascular senescence and its subsequent inflammatory response. Through a comprehensive analysis, we aim to elucidate these knowledge gaps by exploring the network of epigenetic alterations that contribute to vascular senescence. In addition, we describe the consequent inflammatory cascades triggered by these epigenetic modifications. Finally, we explore translational applications involving biomarkers of vascular senescence and the emerging field of senotherapy targeting this biological process.
Collapse
Affiliation(s)
- Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Diego De Leon-Oliva
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Diego Liviu Boaru
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Patricia De Castro-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Cielo Garcia-Montero
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Silvestra Barrena-Blázquez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Joaquin García-García
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Melchor Alvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), Madrid, Spain
- Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine (CIBEREHD), University Hospital Príncipe de Asturias, Alcala deHenares, Spain
| | - Laura Lopez-Gonzalez
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
| | - Raul Diaz-Pedrero
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Department of General and Digestive Surgery, General and Digestive Surgery, Príncipe de Asturias Universitary Hospital, Alcala deHenares, Spain
| | - Luis G. Guijarro
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), Madrid, Spain
- Department of General and Digestive Surgery, General and Digestive Surgery, Príncipe de Asturias Universitary Hospital, Alcala deHenares, Spain
- Unit of Biochemistry and Molecular Biology, Department of System Biology (CIBEREHD), University of Alcalá, Alcala deHenares, Spain
| | - Miguel A. Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala deHenares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), Madrid, Spain
- Cancer Registry and Pathology Department, Principe de Asturias University Hospital, Alcala deHenares, Spain
| |
Collapse
|
11
|
Wang Z, Zhang N, Lin P, Xing Y, Yang N. Recent advances in the treatment and delivery system of diabetic retinopathy. Front Endocrinol (Lausanne) 2024; 15:1347864. [PMID: 38425757 PMCID: PMC10902204 DOI: 10.3389/fendo.2024.1347864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 01/23/2024] [Indexed: 03/02/2024] Open
Abstract
Diabetic retinopathy (DR) is a highly tissue-specific neurovascular complication of type 1 and type 2 diabetes mellitus and is among the leading causes of blindness worldwide. Pathophysiological changes in DR encompass neurodegeneration, inflammation, and oxidative stress. Current treatments for DR, including anti-vascular endothelial growth factor, steroids, laser photocoagulation, and vitrectomy have limitations and adverse reactions, necessitating the exploration of novel treatment strategies. This review aims to summarize the current pathophysiology, therapeutic approaches, and available drug-delivery methods for treating DR, and discuss their respective development potentials. Recent research indicates the efficacy of novel receptor inhibitors and agonists, such as aldose reductase inhibitors, angiotensin-converting enzyme inhibitors, peroxisome proliferator-activated receptor alpha agonists, and novel drugs in delaying DR. Furthermore, with continuous advancements in nanotechnology, a new form of drug delivery has been developed that can address certain limitations of clinical drug therapy, such as low solubility and poor penetration. This review serves as a theoretical foundation for future research on DR treatment. While highlighting promising therapeutic targets, it underscores the need for continuous exploration to enhance our understanding of DR pathogenesis. The limitations of current treatments and the potential for future advancements emphasize the importance of ongoing research in this field.
Collapse
Affiliation(s)
| | | | | | - Yiqiao Xing
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ning Yang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
12
|
Li S, Sun D, Chen S, Zhang S, Gu Q, Shen Y, Xu L, Xu X, Wei F, Wang N. UCP2-SIRT3 Signaling Relieved Hyperglycemia-Induced Oxidative Stress and Senescence in Diabetic Retinopathy. Invest Ophthalmol Vis Sci 2024; 65:14. [PMID: 38175638 PMCID: PMC10774691 DOI: 10.1167/iovs.65.1.14] [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: 05/04/2023] [Accepted: 11/09/2023] [Indexed: 01/05/2024] Open
Abstract
Purpose Diabetic retinopathy (DR) is one of the most common reasons for blindness. uncoupling protein 2 (UCP2), an uncoupling protein located in mitochondria, has been reported to be related to metabolic and vascular diseases. This research aimed to illustrate the function and mechanism of UCP2 in the pathogenesis of DR. Methods Human epiretinal membranes were collected to investigate the expression of UCP2 by quantitative real-time polymerase chain reaction (qRT-PCR) and immunofluorescence. Primary human retinal microvascular endothelial cells (HRECs) were cultured in high glucose (HG) to establish an in vitro cell model for DR. Flow cytometry analysis was used to measure intracellular reactive oxygen species (ROS). Senescence levels were evaluated by the senescence-associated beta-galactosidase (SA-β-gal) assay, the expression of senescence marker P21, and cell-cycle analysis. Adenovirus-mediated UCP2 overexpression or knockdown and specific inhibitors were administered to investigate the underlying regulatory mechanism. Results Proliferative fibrovascular membranes from patients with DR illustrated the downregulation of UCP2 and sirtuin 3 (SIRT3) by qRT-PCR and immunofluorescence. Persistent hyperglycemia-induced UCP2 downregulation in the progress of DR and adenovirus-mediated UCP2 overexpression protected endothelial cells from hyperglycemia-induced oxidative stress and senescence. Under hyperglycemic conditions, UCP2 overexpression attenuated NAD+ downregulation; hence, it promoted the expression and activity of SIRT3, an NAD+-dependent deacetylase regulating mitochondrial function. 3-TYP, a selective SIRT3 inhibitor, abolished the UCP2-mediated protective effect against oxidative stress and senescence. Conclusions UCP2 overexpression relieved oxidative stress and senescence based on a novel mechanism whereby UCP2 can regulate the NAD+-SIRT3 axis. Targeting oxidative stress and senescence amelioration, UCP2-SIRT3 signaling may serve as a method for the prevention and treatment of DR and other diabetic vascular diseases.
Collapse
Affiliation(s)
- Shenping Li
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai, China
| | - Dandan Sun
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai, China
| | - Shimei Chen
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai, China
| | - Shuchang Zhang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai, China
| | - Qing Gu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Diseases, Shanghai, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
| | - Yinchen Shen
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai, China
- National Clinical Research Center for Eye Diseases, Shanghai, China
| | - Li Xu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai, China
| | - Xun Xu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai, China
- National Clinical Research Center for Eye Diseases, Shanghai, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Fang Wei
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai, China
- National Clinical Research Center for Eye Diseases, Shanghai, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Ning Wang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai, China
- National Clinical Research Center for Eye Diseases, Shanghai, China
| |
Collapse
|
13
|
Bhatnagar A, Ting DSW, Weng CY. Treatment Options for Diabetic Macular Edema. Int Ophthalmol Clin 2024; 64:57-69. [PMID: 38146881 DOI: 10.1097/iio.0000000000000518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
|
14
|
Shafqat A, Khan S, Omer MH, Niaz M, Albalkhi I, AlKattan K, Yaqinuddin A, Tchkonia T, Kirkland JL, Hashmi SK. Cellular senescence in brain aging and cognitive decline. Front Aging Neurosci 2023; 15:1281581. [PMID: 38076538 PMCID: PMC10702235 DOI: 10.3389/fnagi.2023.1281581] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/01/2023] [Indexed: 10/16/2024] Open
Abstract
Cellular senescence is a biological aging hallmark that plays a key role in the development of neurodegenerative diseases. Clinical trials are currently underway to evaluate the effectiveness of senotherapies for these diseases. However, the impact of senescence on brain aging and cognitive decline in the absence of neurodegeneration remains uncertain. Moreover, patient populations like cancer survivors, traumatic brain injury survivors, obese individuals, obstructive sleep apnea patients, and chronic kidney disease patients can suffer age-related brain changes like cognitive decline prematurely, suggesting that they may suffer accelerated senescence in the brain. Understanding the role of senescence in neurocognitive deficits linked to these conditions is crucial, especially considering the rapidly evolving field of senotherapeutics. Such treatments could help alleviate early brain aging in these patients, significantly reducing patient morbidity and healthcare costs. This review provides a translational perspective on how cellular senescence plays a role in brain aging and age-related cognitive decline. We also discuss important caveats surrounding mainstream senotherapies like senolytics and senomorphics, and present emerging evidence of hyperbaric oxygen therapy and immune-directed therapies as viable modalities for reducing senescent cell burden.
Collapse
Affiliation(s)
- Areez Shafqat
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | | | - Mohamed H. Omer
- School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Mahnoor Niaz
- Medical College, Aga Khan University, Karachi, Pakistan
| | | | - Khaled AlKattan
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | | | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - James L. Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - Shahrukh K. Hashmi
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
- Clinical Affairs, Khalifa University, Abu Dhabi, United Arab Emirates
- Department of Medicine, SSMC, Abu Dhabi, United Arab Emirates
| |
Collapse
|
15
|
Zhang H, Zhou H, Shen X, Lin X, Zhang Y, Sun Y, Zhou Y, Zhang L, Zhang D. The role of cellular senescence in metabolic diseases and the potential for senotherapeutic interventions. Front Cell Dev Biol 2023; 11:1276707. [PMID: 37868908 PMCID: PMC10587568 DOI: 10.3389/fcell.2023.1276707] [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: 08/12/2023] [Accepted: 09/18/2023] [Indexed: 10/24/2023] Open
Abstract
Cellular senescence represents an irreversible state of cell cycle arrest induced by various stimuli strongly associated with aging and several chronic ailments. In recent years, studies have increasingly suggested that the accumulation of senescent cells is an important contributor to the decline of organ metabolism, ultimately resulting in metabolic diseases. Conversely, the elimination of senescent cells can alleviate or postpone the onset and progression of metabolic diseases. Thus, a close relationship between senescent cells and metabolic diseases is found, and targeting senescent cells has emerged as an alternative therapy for the treatment of metabolic diseases. In this review, we summarize the role of cellular senescence in metabolic diseases, explore relevant therapeutic strategies for metabolic diseases by removing senescent cells, and provide new insights into the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Huantong Zhang
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Han Zhou
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Xin Shen
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Xingchen Lin
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Yuke Zhang
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Yiyi Sun
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Yi Zhou
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Lei Zhang
- School of Economy and Management, Zhejiang Sci-Tech University, Hangzhou, China
- Taizhou Hospital of Zhejiang Province, Zhejiang University, Taizhou, China
| | - Dayong Zhang
- School of Medicine, Hangzhou City University, Hangzhou, China
| |
Collapse
|
16
|
Bang E, Hwangbo H, Kim MY, Ji SY, Kim DH, Shim JH, Moon SK, Kim GY, Cheong J, Choi YH. Urban aerosol particulate matter promotes mitochondrial oxidative stress-induced cellular senescence in human retinal pigment epithelial ARPE-19 cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 102:104211. [PMID: 37423393 DOI: 10.1016/j.etap.2023.104211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/27/2023] [Accepted: 07/03/2023] [Indexed: 07/11/2023]
Abstract
Environmental exposure to urban particulate matter (UPM) is a serious health concern worldwide. Although several studies have linked UPM to ocular diseases, no study has reported effects of UPM exposure on senescence in retinal cells. Therefore, this study aimed to investigate the effects of UPM on senescence and regulatory signaling in human retinal pigment epithelial ARPE-19 cells. Our study demonstrated that UPM significantly promoted senescence, with increased senescence-associated β-galactosidase activity. Moreover, both mRNA and protein levels of senescence markers (p16 and p21) and the senescence-associated secretory phenotype, including IL-1β, matrix metalloproteinase-1, and -3 were upregulated. Notably, UPM increased mitochondrial reactive oxygen species-dependent nuclear factor-kappa B (NF-κB) activation during senescence. In contrast, use of NF-κB inhibitor Bay 11-7082 reduced the level of senescence markers. Taken together, our results provide the first in vitro preliminary evidence that UPM induces senescence by promoting mitochondrial oxidative stress-mediated NF-κB activation in ARPE-19 cells.
Collapse
Affiliation(s)
- EunJin Bang
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Republic of Korea; Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea
| | - Hyun Hwangbo
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Republic of Korea; Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea
| | - Min Yeong Kim
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Republic of Korea; Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea
| | - Seon Yeong Ji
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Republic of Korea; Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea
| | - Da Hye Kim
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Republic of Korea; Department of Molecular Biology, Pusan National University, Busan 46241, Republic of Korea
| | - Jung-Hyun Shim
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea
| | - Sung-Kwon Moon
- Department of Food and Nutrition, Chung-Ang University, Ansung 17546, Republic of Korea
| | - Gi-Young Kim
- Department of Marine Life Science, Jeju National University, Jeju 63243, Republic of Korea
| | - Jaehun Cheong
- Department of Molecular Biology, Pusan National University, Busan 46241, Republic of Korea
| | - Yung Hyun Choi
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Republic of Korea; Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea.
| |
Collapse
|
17
|
Rzepka Z, Rok J, Kowalska J, Banach K, Wrześniok D. Cobalamin Deficiency May Induce Astrosenescence-An In Vitro Study. Cells 2022; 11:3408. [PMID: 36359805 PMCID: PMC9655094 DOI: 10.3390/cells11213408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/19/2022] [Accepted: 10/26/2022] [Indexed: 11/30/2022] Open
Abstract
Cobalamin (vitamin B12) deficiency is one of the major factors causing degenerative changes in the nervous system and, thus, various neurological and psychiatric symptoms. The underlying cellular mechanism of this phenomenon is not yet fully understood. An accumulation of senescent astrocytes has been shown to contribute to a wide range of pathologies of the nervous system, including neurodegenerative disorders. This study aimed to investigate whether cobalamin deficiency triggers astrosenescence. After inducing cobalamin deficiency in normal human astrocytes in vitro, we examined biomarkers of cellular senescence: SA-β-gal, p16INK4A, and p21Waf1/Cip1 and performed cell nuclei measurements. The obtained results may contribute to an increase in the knowledge of the cellular effects of cobalamin deficiency in the context of astrocytes. In addition, the presented data suggest a potential causative agent of astrosenescence that has not been proven to date.
Collapse
Affiliation(s)
- Zuzanna Rzepka
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 4 Jagiellońska Str., 41-200 Sosnowiec, Poland
| | | | | | | | | |
Collapse
|