151
|
Jiang A, Liu Y, Zhu B, Fang Y, Qu L, Yang Q, Luo P, Cai C, Wang L. SPCS, a Novel Classifier System Based on Senescence Axis Regulators Reveals Tumor Microenvironment Heterogeneity and Guides Frontline Therapy for Clear Cell Renal Carcinoma. Clin Genitourin Cancer 2024; 22:497-513. [PMID: 38245436 DOI: 10.1016/j.clgc.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 01/03/2024] [Accepted: 01/06/2024] [Indexed: 01/22/2024]
Abstract
RATIONALE The emerging evidence suggested that senescence regulator genes were involved in multi cancers, which may be utilized as new targets for cancers. However, the dysregulation and clinical impact of senescence regulator genes in clear cell renal cell cancer (ccRCC) were still in foggy. METHODS Using multiomics data from TCGA-KIRC and other datasets, we comprehensively investigated the function of senescence regulator genes in ccRCC. ccRCC patients could be remodeled into 2 significant different groups basing on senescence regulators expression: senescence-pattern cancer subtype1 (SPCS1) and subtype2 (SPCS2). We further explored clinical characteristics, functional analysis, tumor immune microenvironment, immunotherapy response, genomic mutation and drug sensitivity between the 2 subtypes. Besides, senescence-pattern related risk model was established to determine the patient's prognosis of ccRCC. Finally, the overview of MECP2 function was investigated in multi cancers. RESULTS ccRCC patients could be divided into SPCS1 (normal aging group) and SPCS2 (Aging disorder group). The 2 subtypes showed significant different clinical characteristics and biological process in ccRCC. SPCS2, an aggressive subtype, comprised higher clinical stage and worse prognosis of ccRCC patients. SPCS2 subtype indicated activated oncogenic signaling pathway and metabolic signatures to prompt cancer expansion. SPCS2 subgroup owned immunocompromised status, which induced immune dysfunction and low ICI therapy response. The genome-copy numbers of SPCS2, including arm-gain and arm-loss was significantly more frequent than SPCS1. In addition, the 2 subtypes argue contrasting drug sensitivity profiles in clinical specimens and matched cell lines. Finally, we constructed a prognostic risk model consisted of each subtype's leading biomarkers, which exerted a satisfied performance for ccRCC patients. CONCLUSION Senescence regulator-related signature could modify functional pathways and tumor immune microenvironment by genome mutation and pathway interaction. Senescence regulator-related molecular subtype strengthen the understanding of ccRCC' characterization and guide clinical treatment. Targeting senescence regulators may be regard as a proper way in ccRCC.
Collapse
Affiliation(s)
- Aimin Jiang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Ying Liu
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Baohua Zhu
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Yu Fang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Le Qu
- Department of Urology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Qiwei Yang
- Depanrtment of Urology, The Third Affiliated Hospital of Naval Military Medical University (Eastern Hepatobiliary Surgery Hospital), Shanghai, China; Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Chen Cai
- Department of Special Clinic, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China.
| | - Linhui Wang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China.
| |
Collapse
|
152
|
Li T, Hasan MN, Gu L. Bile acids regulation of cellular stress responses in liver physiology and diseases. EGASTROENTEROLOGY 2024; 2:e100074. [PMID: 39027418 PMCID: PMC11257078 DOI: 10.1136/egastro-2024-100074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Bile acids are physiological detergents and signalling molecules that are critically implicated in liver health and diseases. Dysregulation of bile acid homeostasis alters cell function and causes cell injury in chronic liver diseases. Therapeutic agents targeting bile acid synthesis, transport and signalling hold great potential for treatment of chronic liver diseases. The broad cellular and physiological impacts of pharmacological manipulations of bile acid metabolism are still incompletely understood. Recent research has discovered new links of bile acid signalling to the regulation of autophagy and lysosome biology, redox homeostasis and endoplasmic reticulum stress. These are well-conserved mechanisms that allow cells to adapt to nutrient and organelle stresses and play critical roles in maintaining cellular integrity and promoting survival. However, dysregulation of these cellular pathways is often observed in chronic liver diseases, which exacerbates cellular dysfunction to contribute to disease pathogenesis. Therefore, identification of these novel links has significantly advanced our knowledge of bile acid biology and physiology, which is needed to understand the contributions of bile acid dysregulation in disease pathogenesis, establish bile acids as diagnostic markers and develop bile acid-based pharmacological interventions. In this review, we will first discuss the roles of bile acid dysregulation in the pathogenesis of chronic liver diseases, and then discuss the recent findings on the crosstalk of bile acid signalling and cellular stress responses. Future investigations are needed to better define the roles of these crosstalks in regulating cellular function and disease processes.
Collapse
Affiliation(s)
- Tiangang Li
- Department of Biochemistry and Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Mohammad Nazmul Hasan
- Department of Biochemistry and Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Lijie Gu
- Department of Biochemistry and Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
153
|
Sato S, Ogawa Y, Shimizu E, Asai K, Okazaki T, Rusch R, Hirayama M, Shimmura S, Negishi K, Tsubota K. Cellular senescence promotes meibomian gland dysfunction in a chronic graft-versus-host disease mouse model. Ocul Surf 2024; 32:198-210. [PMID: 38499288 DOI: 10.1016/j.jtos.2024.03.006] [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: 09/27/2023] [Revised: 01/27/2024] [Accepted: 03/12/2024] [Indexed: 03/20/2024]
Abstract
PURPOSE Aging is a well-established risk factor for meibomian gland dysfunction (MGD). We previously reported an accelerated cellular senescence phenomenon in the lacrimal glands of a murine model of chronic graft-versus-host disease (cGVHD). Herein, we aimed to elucidate the relationship between cellular senescence and MGD in cGVHD mice, utilizing the senolytic agent ABT-263. METHODS A cGVHD mouse model was established through allogeneic bone marrow transplantation (BMT) from B10.D2 to BALB/c mice. Subsequently, cGVHD mice were treated with either ABT-263 or vehicle. The eyelids of recipients were analyzed at 4-week intervals post-BMT in both groups. RESULTS Meibomian gland (MG) area was significantly smaller in cGVHD mice than in syngeneic control mice. ABT-263-treated mice retained a significantly larger MG area than their vehicle-treated counterparts. Pathological and immunohistochemical examinations revealed significant reductions in eyelid tissue inflammation and pathological fibrosis in the ABT-263 group compared to that in the vehicle-treated group. Additionally, expression of DNA damage markers, senescent cell markers, and senescence-associated secretory phenotype (SASP) factors was elevated in the eyelids of cGVHD mice compared with that in syngeneic mice. The expression of these cellular senescence-associated molecules was considerably suppressed in ABT-263-treated eyelids compared to that in vehicle-treated ones. CONCLUSIONS Cellular senescence, along with expression of SASP factors, exhibited increased activity in the eyelids, particularly in the MGs of cGVHD mice. ABT-263 mitigated the severity of MGD. These findings highlight the potential of targeting cellular senescence as an effective approach for MGD treatment in cGVHD.
Collapse
Affiliation(s)
- Shinri Sato
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan.
| | - Yoko Ogawa
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan.
| | - Eisuke Shimizu
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Kazuki Asai
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Takahiro Okazaki
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Robert Rusch
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Masatoshi Hirayama
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Shigeto Shimmura
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan; Fujita Medical Innovation Center Tokyo, Fujita Health University, Tokyo, Japan
| | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | | |
Collapse
|
154
|
Ma Y, Li S, Ye S, Luo S, Wei L, Su Y, Zeng Y, Shi Y, Bian H, Xiao F. The role of miR-222-2p in exosomes secreted by hexavalent chromium-induced premature senescent hepatocytes as a SASP component. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 346:123535. [PMID: 38365080 DOI: 10.1016/j.envpol.2024.123535] [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: 05/09/2023] [Revised: 01/21/2024] [Accepted: 02/07/2024] [Indexed: 02/18/2024]
Abstract
With the development of world industrialization, the environmental pollution of hexavalent chromium [Cr(VI)] is becoming an increasingly serious problem. In particular, the mechanisms by which long-term and low-dose exposure to Cr(VI) leading the development of related cancers are not well understood. As senescent cells gradually lose their ability to proliferate and divide, they will not be malignantly transformed. However, Senescence-associated secretory phenotype (SASP) released by senescent cells into the cellular microenvironment can act on neighboring cells. Since SASP has a bidirectional regulatory role in the malignant transformation of cells. Hence, It is very necessary to identified the composition and function of SASP which secreted by Cr(VI) induced senescent L02 hepatocytes (S-L02). Exosomes, a vesicle-like substances released extracellularly after the fusion of intracellular multivesicular bodies with cell membrane, are important components of SASP and contain a large number of microRNAs (miRNAs). By establishing Cr(VI)-induced S-L02 model, we collected the exosomes from the supernatants of S-L02 and L02 culture medium respectively, and screened out the highly expressed miRNAs in the exosomes of S-L02, namely the new SASP components. Among them, the increase of miR-222-5p was the most significant. It was validated that as SASP, miR-222-5p can inhibit the proliferation of L02 and S-L02 hepatocytes and at the same time accelerate the proliferation and migration ability of HCC cells. Further mechanistic studies revealed that miR-222-5p attenuated the regulatory effect of protein phosphatase 2A subunit B isoform R2-α (PPP2R2A) on Akt via repressing its target gene PPP2R2A, causing reduced expressions of forkhead box O3 (FOXO3a), p27 and p21, and finally increasing the proliferation of HCC cells after diminishing the negative regulation of on cell cycle. This study certainly provides valuable laboratory evidence as well as potential therapeutic targets for the prevention and further personalized treatment of Cr(VI)-associated cancers.
Collapse
Affiliation(s)
- Yu Ma
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha, 410078, China
| | - Siwen Li
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha, 410078, China
| | - Shuzi Ye
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha, 410078, China
| | - Sijia Luo
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha, 410078, China
| | - Lai Wei
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha, 410078, China
| | - Ying Su
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha, 410078, China
| | - Yuan Zeng
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha, 410078, China
| | - Yan Shi
- Institute of Environmental Science and Engineering, School of Metallurgy and Environment, Central South University, 410083, Changsha, China; National Engineering Research Center for Heavy Metals Pollution Control and Treatment, 410083, Changsha, China
| | - Huanfeng Bian
- Shajing Sub-Center of Public Health Service, Bao'an District, 518125, Shenzhen, Guangdong, China
| | - Fang Xiao
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha, 410078, China.
| |
Collapse
|
155
|
Qiao QF, Wang LQ, Yu DE, Li N, Xu QJ, Zhou YJ. Effect of beta-cypermethrin on the reproductive capacity of female mice in advanced age. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 107:104410. [PMID: 38423490 DOI: 10.1016/j.etap.2024.104410] [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: 01/30/2023] [Revised: 02/07/2024] [Accepted: 02/26/2024] [Indexed: 03/02/2024]
Abstract
The aim of the present study was to investigate whether exposure to pesticides beta-cypermethrin (β-CYP) harms the reproductive capacity of advanced-age female mice. The results evidenced that peri-implantation β-CYP exposure significantly reduced the number of fetuses per advanced-age female in the first litter, and the number and weight of implantation sites. The levels of decidualization markers were significantly reduced in β-CYP-administered advanced-age mice. Lower expression of Pcna, Cdk6, Foxo1, Ki67, and p62 protein and mRNA was found in the decidua of β-CYP-treated advanced-age mice. The levels of Bax, cleaved caspase-3, Lc3a/b, Atg, mTOR, and p-mTOR protein, and the ratio of p-mTOR/mTOR protein expression were clearly downregulated by peri-implantation β-CYP exposure. These results indicated that peri-implantation β-CYP exposure may elevate the decline in reproductive capacity of early pregnant mice in advanced age.
Collapse
Affiliation(s)
- Qian-Feng Qiao
- International School of Public Health and One Health, Hainan Medical University, Hainan Province 571199, People's Republic of China
| | - Li-Qing Wang
- Maternal, Child and Adolescent Health, International School of Public Health and One Health, Hainan Medical University, Hainan Province 571199, People's Republic of China
| | - De-E Yu
- International School of Public Health and One Health, Hainan Medical University, Hainan Province 571199, People's Republic of China
| | - Na Li
- International School of Public Health and One Health, Hainan Medical University, Hainan Province 571199, People's Republic of China
| | - Qiong-Jun Xu
- International School of Public Health and One Health, Hainan Medical University, Hainan Province 571199, People's Republic of China
| | - Yong-Jiang Zhou
- Heinz Mehlhorn Academician Workstation, Maternal, Child and Adolescent Health, International School of Public Health and One Health, Hainan Medical University, Hainan Province 571199, People's Republic of China.
| |
Collapse
|
156
|
Liu Z, Zhang Y, Li D, Fu J. Cellular senescence in chronic lung diseases from newborns to the elderly: An update literature review. Biomed Pharmacother 2024; 173:116463. [PMID: 38503240 DOI: 10.1016/j.biopha.2024.116463] [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: 12/06/2023] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 03/21/2024] Open
Abstract
The role of cellular senescence in age-related diseases has been fully recognized. In various age-related-chronic lung diseases, the function of alveolar epithelial cells (AECs) is impaired and alveolar regeneration disorders, especially in bronchopulmonary dysplasia,pulmonary fibrosis (PF), chronic obstructive pulmonary disease (COPD), cancer, etc. Except for age-related-chronic lung diseases, an increasing number of studies are exploring the role of cellular senescence in developmental chronic lung diseases, which typically originate in childhood and even in the neonatal period. This review provides an overview of cellular senescence and lung diseases from newborns to the elderly, attempting to draw attention to the relationship between cellular senescence and developmental lung diseases.
Collapse
Affiliation(s)
- Ziyun Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Yiqi Zhang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Danni Li
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China.
| | - Jianhua Fu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China.
| |
Collapse
|
157
|
Ali I, Zhang H, Zaidi SAA, Zhou G. Understanding the intricacies of cellular senescence in atherosclerosis: Mechanisms and therapeutic implications. Ageing Res Rev 2024; 96:102273. [PMID: 38492810 DOI: 10.1016/j.arr.2024.102273] [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: 11/14/2023] [Revised: 01/16/2024] [Accepted: 03/13/2024] [Indexed: 03/18/2024]
Abstract
Cardiovascular disease is currently the largest cause of mortality and disability globally, surpassing communicable diseases, and atherosclerosis is the main contributor to this epidemic. Aging is intimately linked to atherosclerosis development and progression, however, the mechanism of aging in atherosclerosis is not well known. To emphasize the significant research on the involvement of senescent cells in atherosclerosis, we begin by outlining compelling evidence that indicates various types of senescent cells and SASP factors linked to atherosclerotic phenotypes. We subsequently provide a comprehensive summary of the existing knowledge, shedding light on the intricate mechanisms through which cellular senescence contributes to the pathogenesis of atherosclerosis. Further, we cover that senescence can be identified by both structural changes and several senescence-associated biomarkers. Finally, we discuss that preventing accelerated cellular senescence represents an important therapeutic potential, as permanent changes may occur in advanced atherosclerosis. Together, the review summarizes the relationship between cellular senescence and atherosclerosis, and inspects the molecular knowledge, and potential clinical significance of senescent cells in developing senescent-based therapy, thus providing crucial insights into their biology and potential therapeutic exploration.
Collapse
Affiliation(s)
- Ilyas Ali
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, PR China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, PR China
| | - Hongliang Zhang
- Shenzhen University General Hospital, Shenzhen University, Shenzhen 518060, PR China
| | - Syed Aqib Ali Zaidi
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, PR China
| | - Guangqian Zhou
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen 518060, PR China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, PR China.
| |
Collapse
|
158
|
Kuga T, Chiba A, Murayama G, Hosomi K, Nakagawa T, Yahagi Y, Noto D, Kusaoi M, Kawano F, Yamaji K, Tamura N, Miyake S. Enhanced GATA4 expression in senescent systemic lupus erythematosus monocytes promotes high levels of IFNα production. Front Immunol 2024; 15:1320444. [PMID: 38605949 PMCID: PMC11007064 DOI: 10.3389/fimmu.2024.1320444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 03/11/2024] [Indexed: 04/13/2024] Open
Abstract
Enhanced interferon α (IFNα) production has been implicated in the pathogenesis of systemic lupus erythematosus (SLE). We previously reported IFNα production by monocytes upon activation of the stimulator of IFN genes (STING) pathway was enhanced in patients with SLE. We investigated the mechanism of enhanced IFNα production in SLE monocytes. Monocytes enriched from the peripheral blood of SLE patients and healthy controls (HC) were stimulated with 2'3'-cyclic GAMP (2'3'-cGAMP), a ligand of STING. IFNα positive/negative cells were FACS-sorted for RNA-sequencing analysis. Gene expression in untreated and 2'3'-cGAMP-stimulated SLE and HC monocytes was quantified by real-time PCR. The effect of GATA binding protein 4 (GATA4) on IFNα production was investigated by overexpressing GATA4 in monocytic U937 cells by vector transfection. Chromatin immunoprecipitation was performed to identify GATA4 binding target genes in U937 cells stimulated with 2'3'-cGAMP. Differentially expressed gene analysis of cGAS-STING stimulated SLE and HC monocytes revealed the enrichment of gene sets related to cellular senescence in SLE. CDKN2A, a marker gene of cellular senescence, was upregulated in SLE monocytes at steady state, and its expression was further enhanced upon STING stimulation. GATA4 expression was upregulated in IFNα-positive SLE monocytes. Overexpression of GATA4 enhanced IFNα production in U937 cells. GATA4 bound to the enhancer region of IFIT family genes and promoted the expressions of IFIT1, IFIT2, and IFIT3, which promote type I IFN induction. SLE monocytes with accelerated cellular senescence produced high levels of IFNα related to GATA4 expression upon activation of the cGAS-STING pathway.
Collapse
Affiliation(s)
- Taiga Kuga
- Department of Immunology, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
- Department of Internal Medicine and Rheumatology, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Asako Chiba
- Department of Immunology, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Goh Murayama
- Department of Internal Medicine and Rheumatology, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Kosuke Hosomi
- Department of Immunology, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Tomoya Nakagawa
- Department of Immunology, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Yoshiyuki Yahagi
- Department of Immunology, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
- Department of Internal Medicine and Rheumatology, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Daisuke Noto
- Department of Immunology, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Makio Kusaoi
- Department of Internal Medicine and Rheumatology, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Fuminori Kawano
- Graduate School of Health Sciences, Matsumoto University, Matsumoto, Nagano, Japan
| | - Ken Yamaji
- Department of Internal Medicine and Rheumatology, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Naoto Tamura
- Department of Internal Medicine and Rheumatology, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Sachiko Miyake
- Department of Immunology, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
159
|
Melones-Herrero J, Alcalá S, Ruiz-Cañas L, Benítez-Buelga C, Batres-Ramos S, Calés C, Lorenzo O, Perona R, Quiroga AG, Sainz B, Sánchez-Pérez I. Platinum iodido drugs show potential anti-tumor activity, affecting cancer cell metabolism and inducing ROS and senescence in gastrointestinal cancer cells. Commun Biol 2024; 7:353. [PMID: 38519773 PMCID: PMC10959927 DOI: 10.1038/s42003-024-06052-5] [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: 05/24/2023] [Accepted: 03/14/2024] [Indexed: 03/25/2024] Open
Abstract
Cisplatin-based chemotherapy has associated clinical disadvantages, such as high toxicity and resistance. Thus, the development of new antitumor metallodrugs able to overcome different clinical barriers is a public healthcare priority. Here, we studied the mechanism of action of the isomers trans and cis-[PtI2(isopropylamine)2] (I5 and I6, respectively) against gastrointestinal cancer cells. We demonstrate that I5 and I6 modulate mitochondrial metabolism, decreasing OXPHOS activity and negatively affecting ATP-linked oxygen consumption rate. Consequently, I5 and I6 generated Reactive Oxygen Species (ROS), provoking oxidative damage and eventually the induction of senescence. Thus, herein we propose a loop with three interconnected processes modulated by these iodido agents: (i) mitochondrial dysfunction and metabolic disruptions; (ii) ROS generation and oxidative damage; and (iii) cellular senescence. Functionally, I5 reduces cancer cell clonogenicity and tumor growth in a pancreatic xenograft model without systemic toxicity, highlighting a potential anticancer complex that warrants additional pre-clinical studies.
Collapse
Affiliation(s)
- Jorge Melones-Herrero
- Department of Biochemistry. School of Medicine, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Instituto de Investigaciones Biomédicas "Sols-Morreale" IIBM-CSIC-UAM, Madrid, Spain
- Biomarkers and Personalized Approach to Cancer (BioPAC) Group. Area 3 Cancer -Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Sonia Alcalá
- Department of Biochemistry. School of Medicine, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Instituto de Investigaciones Biomédicas "Sols-Morreale" IIBM-CSIC-UAM, Madrid, Spain
- Biomarkers and Personalized Approach to Cancer (BioPAC) Group. Area 3 Cancer -Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Laura Ruiz-Cañas
- Department of Biochemistry. School of Medicine, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Instituto de Investigaciones Biomédicas "Sols-Morreale" IIBM-CSIC-UAM, Madrid, Spain
- Biomarkers and Personalized Approach to Cancer (BioPAC) Group. Area 3 Cancer -Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Carlos Benítez-Buelga
- Instituto de Investigaciones Biomédicas "Sols-Morreale" IIBM-CSIC-UAM, Madrid, Spain
| | - Sandra Batres-Ramos
- Department of Biochemistry. School of Medicine, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Instituto de Investigaciones Biomédicas "Sols-Morreale" IIBM-CSIC-UAM, Madrid, Spain
- Biomarkers and Personalized Approach to Cancer (BioPAC) Group. Area 3 Cancer -Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Carmela Calés
- Department of Biochemistry. School of Medicine, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Instituto de Investigaciones Biomédicas "Sols-Morreale" IIBM-CSIC-UAM, Madrid, Spain
| | - Oscar Lorenzo
- Laboratory of Diabetes and Vascular Pathology, Instituto de Investigaciones Sanitarias-Fundación Jimenez Díaz, CIBERDEM, UAM, Madrid, Spain
| | - Rosario Perona
- Instituto de Investigaciones Biomédicas "Sols-Morreale" IIBM-CSIC-UAM, Madrid, Spain
- Centro de Investigación Biomédica en Red, Área Rare Diseases, CIBERER, ISCIII, Madrid, Spain
- Instituto de Salud Carlos III, Madrid, Spain
| | - Adoración G Quiroga
- Department of Inorganic Chemistry, School of Sciences, IAdChem, UAM, Madrid, Spain
| | - Bruno Sainz
- Instituto de Investigaciones Biomédicas "Sols-Morreale" IIBM-CSIC-UAM, Madrid, Spain.
- Biomarkers and Personalized Approach to Cancer (BioPAC) Group. Area 3 Cancer -Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.
- Centro de Investigación Biomédica en Red, Área Cáncer, CIBERONC, ISCIII, Madrid, Spain.
| | - Isabel Sánchez-Pérez
- Department of Biochemistry. School of Medicine, Universidad Autónoma de Madrid (UAM), Madrid, Spain.
- Instituto de Investigaciones Biomédicas "Sols-Morreale" IIBM-CSIC-UAM, Madrid, Spain.
- Biomarkers and Personalized Approach to Cancer (BioPAC) Group. Area 3 Cancer -Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.
- Centro de Investigación Biomédica en Red, Área Rare Diseases, CIBERER, ISCIII, Madrid, Spain.
- Unidad Asociada de Biomedicina UCLM-CSIC, Madrid, Spain.
| |
Collapse
|
160
|
Shreeya T, Ansari MS, Kumar P, Saifi M, Shati AA, Alfaifi MY, Elbehairi SEI. Senescence: A DNA damage response and its role in aging and Neurodegenerative Diseases. FRONTIERS IN AGING 2024; 4:1292053. [PMID: 38596783 PMCID: PMC11002673 DOI: 10.3389/fragi.2023.1292053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 11/09/2023] [Indexed: 04/11/2024]
Abstract
Senescence is a complicated, multi-factorial, irreversible cell cycle halt that has a tumor-suppressing effect in addition to being a significant factor in aging and neurological diseases. Damaged DNA, neuroinflammation, oxidative stress and disrupted proteostasis are a few of the factors that cause senescence. Senescence is triggered by DNA damage which initiates DNA damage response. The DNA damage response, which includes the formation of DNA damage foci containing activated H2AX, which is a key factor in cellular senescence, is provoked by a double strand DNA break. Oxidative stress impairs cognition, inhibits neurogenesis, and has an accelerated aging effect. Senescent cells generate pro-inflammatory mediators known as senescence-associated secretory phenotype (SASP). These pro-inflammatory cytokines and chemokines have an impact on neuroinflammation, neuronal death, and cell proliferation. While it is tempting to think of neurodegenerative diseases as manifestations of accelerated aging and senescence, this review will present information on brain ageing and neurodegeneration as a result of senescence and DNA damage response.
Collapse
Affiliation(s)
- Tejal Shreeya
- Institute of Biophysics, Biological Research Center, Szeged, Hungary
- Doctoral School of Theoretical Medicine, University of Szeged, Szeged, Hungary
| | - Mohd Saifullah Ansari
- Institute of Genetics, Biological Research Center, Szeged, Hungary
- Doctoral School of Biology, University of Szeged, Szeged, Hungary
| | - Prabhat Kumar
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Centre for Neuroscience, University of Pécs, Pécs, Hungary
| | | | - Ali A. Shati
- Biology Department, Faculty of Science, King Khalid University, Abha, Saudi Arabia
| | - Mohammad Y. Alfaifi
- Biology Department, Faculty of Science, King Khalid University, Abha, Saudi Arabia
| | | |
Collapse
|
161
|
Luo Y, Zhang L, Su N, Liu L, Zhao T. YME1L-mediated mitophagy protects renal tubular cells against cellular senescence under diabetic conditions. Biol Res 2024; 57:10. [PMID: 38494498 PMCID: PMC10946153 DOI: 10.1186/s40659-024-00487-0] [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: 08/19/2023] [Accepted: 02/27/2024] [Indexed: 03/19/2024] Open
Abstract
BACKGROUND The senescence of renal tubular epithelial cells (RTECs) is crucial in the progression of diabetic kidney disease (DKD). Accumulating evidence suggests a close association between insufficient mitophagy and RTEC senescence. Yeast mitochondrial escape 1-like 1 (YME1L), an inner mitochondrial membrane metalloprotease, maintains mitochondrial integrity. Its functions in DKD remain unclear. Here, we investigated whether YME1L can prevent the progression of DKD by regulating mitophagy and cellular senescence. METHODS We analyzed YME1L expression in renal tubules of DKD patients and mice, explored transcriptomic changes associated with YME1L overexpression in RTECs, and assessed its impact on RTEC senescence and renal dysfunction using an HFD/STZ-induced DKD mouse model. Tubule-specific overexpression of YME1L was achieved through the use of recombinant adeno-associated virus 2/9 (rAAV 2/9). We conducted both in vivo and in vitro experiments to evaluate the effects of YME1L overexpression on mitophagy and mitochondrial function. Furthermore, we performed LC-MS/MS analysis to identify potential protein interactions involving YME1L and elucidate the underlying mechanisms. RESULTS Our findings revealed a significant decrease in YME1L expression in the renal tubules of DKD patients and mice. However, tubule-specific overexpression of YME1L significantly alleviated RTEC senescence and renal dysfunction in the HFD/STZ-induced DKD mouse model. Moreover, YME1L overexpression exhibited positive effects on enhancing mitophagy and improving mitochondrial function both in vivo and in vitro. Mechanistically, our LC-MS/MS analysis uncovered a crucial mitophagy receptor, BCL2-like 13 (BCL2L13), as an interacting partner of YME1L. Furthermore, YME1L was found to promote the phosphorylation of BCL2L13, highlighting its role in regulating mitophagy. CONCLUSIONS This study provides compelling evidence that YME1L plays a critical role in protecting RTECs from cellular senescence and impeding the progression of DKD. Overexpression of YME1L demonstrated significant therapeutic potential by ameliorating both RTEC senescence and renal dysfunction in the DKD mice. Moreover, our findings indicate that YME1L enhances mitophagy and improves mitochondrial function, potentially through its interaction with BCL2L13 and subsequent phosphorylation. These novel insights into the protective mechanisms of YME1L offer a promising strategy for developing therapies targeting DKD.
Collapse
Affiliation(s)
- Yuanyuan Luo
- Department of Endocrinology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
- Department of Endocrinology, Chongqing University Three Gorges Hospital, Chongqing, 404000, China
| | - Lingxiao Zhang
- Department of Endocrinology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
| | - Ning Su
- Department of Hematology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
| | - Lerong Liu
- Department of Endocrinology, Southern Medical University Nanfang Hospital, Guangzhou, 510515, China
| | - Tongfeng Zhao
- Department of Endocrinology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China.
- Department of Endocrinology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 51000, China.
| |
Collapse
|
162
|
Neri F, Takajjart SN, Lerner CA, Desprez PY, Schilling B, Campisi J, Gerencser AA. A Fully-Automated Senescence Test (FAST) for the high-throughput quantification of senescence-associated markers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.22.573123. [PMID: 38187756 PMCID: PMC10769423 DOI: 10.1101/2023.12.22.573123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Cellular senescence is a major driver of aging and age-related diseases. Quantification of senescent cells remains challenging due to the lack of senescence-specific markers and generalist, unbiased methodology. Here, we describe the Fully-Automated Senescence Test (FAST), an image-based method for the high-throughput, single-cell assessment of senescence in cultured cells. FAST quantifies three of the most widely adopted senescence-associated markers for each cell imaged: senescence-associated β-galactosidase activity (SA-β-Gal) using X-Gal, proliferation arrest via lack of 5-ethynyl-2'-deoxyuridine (EdU) incorporation, and enlarged morphology via increased nuclear area. The presented workflow entails microplate image acquisition, image processing, data analysis, and graphing. Standardization was achieved by i) quantifying colorimetric SA-β-Gal via optical density; ii) implementing staining background controls; iii) automating image acquisition, image processing, and data analysis. In addition to the automated threshold-based scoring, a multivariate machine learning approach is provided. We show that FAST accurately quantifies senescence burden and is agnostic to cell type and microscope setup. Moreover, it effectively mitigates false-positive senescence marker staining, a common issue arising from culturing conditions. Using FAST, we compared X-Gal with fluorescent C12FDG live-cell SA-β-Gal staining on the single-cell level. We observed only a modest correlation between the two, indicating that those stains are not trivially interchangeable. Finally, we provide proof of concept that our method is suitable for screening compounds that modify senescence burden. This method will be broadly useful to the aging field by enabling rapid, unbiased, and user-friendly quantification of senescence burden in culture, as well as facilitating large-scale experiments that were previously impractical.
Collapse
Affiliation(s)
- Francesco Neri
- Buck Institute for Research on Aging, Novato, CA, USA
- USC Leonard Davis School of Gerontology, Los Angeles, CA, USA
| | | | | | - Pierre-Yves Desprez
- Buck Institute for Research on Aging, Novato, CA, USA
- California Pacific Medical Center, San Francisco, CA, USA
| | - Birgit Schilling
- Buck Institute for Research on Aging, Novato, CA, USA
- USC Leonard Davis School of Gerontology, Los Angeles, CA, USA
| | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA, USA
- USC Leonard Davis School of Gerontology, Los Angeles, CA, USA
| | | |
Collapse
|
163
|
Yu PJ, Zhou M, Liu Y, Du J. Senescent T Cells in Age-Related Diseases. Aging Dis 2024:AD.2024.0219. [PMID: 38502582 DOI: 10.14336/ad.2024.0219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/18/2024] [Indexed: 03/21/2024] Open
Abstract
Age-induced alterations in human immunity are often considered deleterious and are referred to as immunosenescence. The immune system monitors the number of senescent cells in the body, while immunosenescence may represent the initiation of systemic aging. Immune cells, particularly T cells, are the most impacted and involved in age-related immune function deterioration, making older individuals more prone to different age-related diseases. T-cell senescence can impact the effectiveness of immunotherapies that rely on the immune system's function, including vaccines and adoptive T-cell therapies. The research and practice of using senescent T cells as therapeutic targets to intervene in age-related diseases are in their nascent stages. Therefore, in this review, we summarize recent related literature to investigate the characteristics of senescent T cells as well as their formation mechanisms, relationship with various aging-related diseases, and means of intervention. The primary objective of this article is to explore the prospects and possibilities of therapeutically targeting senescent T cells, serving as a valuable resource for the development of immunotherapy and treatment of age-related diseases.
Collapse
|
164
|
Matveeva K, Vasilieva M, Minskaia E, Rybtsov S, Shevyrev D. T-cell immunity against senescence: potential role and perspectives. Front Immunol 2024; 15:1360109. [PMID: 38504990 PMCID: PMC10948549 DOI: 10.3389/fimmu.2024.1360109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/19/2024] [Indexed: 03/21/2024] Open
Abstract
The development of age-associated diseases is related to the accumulation of senescent cells in the body. These are old non-functional cells with impaired metabolism, which are unable to divide. Such cells are also resistant to programmed cell death and prone to spontaneous production of some inflammatory factors. The accumulation of senescent cells is related to the age-associated dysfunction of organs and tissues as well as chronic inflammation that enhances with age. In the young organism, senescent cells are removed with the innate immunity system. However, the efficiency of this process decreases with age. Nowadays, more and more evidences are accumulating to support the involvement of specific immunity and T-lymphocytes in the fight against senescent cells. It has great physiological importance since the efficient elimination of senescent cells requires a high diversity of antigen-recognizing receptors to cover the entire spectrum of senescent-associated antigens with high precision and specificity. Developing the approaches of T-cell immunity stimulation to generate or amplify a physiological immune response against senescent cells can provide new perspectives to extend active longevity. In this mini-review, the authors summarize the current understanding of the role of T-cell immunity in the fight against senescent cells and discuss the prospects of stimulating adaptive immunity for combating the accumulation of senescent cells that occurs with age.
Collapse
|
165
|
Zhu N, Li Y, Xu M. Beneficial Effects of Small-Molecule Oligopeptides Isolated from Panax Ginseng C. A. Meyer on Cellular Fates in Oxidative Stress-Induced Damaged Human Umbilical Vein Endothelial Cells and PC-12. Int J Mol Sci 2024; 25:2906. [PMID: 38474153 DOI: 10.3390/ijms25052906] [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: 12/29/2023] [Revised: 01/21/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Cell fate instability is a crucial characteristic of aging and appears to contribute to various age-related pathologies. Exploring the connection between bioactive substances and cell fate stability may offer valuable insights into longevity. Therefore, the objective of this study was to investigate the potential beneficial effects of ginseng oligopeptides (GOPs) isolated from Panax ginseng C. A. Meyer at the cellular level. Disruption of homeostasis of human umbilical vein endothelial cells (HUVECs) and PC-12 was achieved by culturing them in the growth medium supplemented with 200 µM of H2O2, and 25, 50, and 100 µg/mL GOPs for 4 h. Then, they were cultured in a H2O2-free growth medium containing different concentration of GOPs. We found that GOP administration retards the oxidative stress-induced cell instability in HUVECs by increasing cell viability, inhibiting the cell cycle arrest, enhancing telomerase (TE) activity, suppressing oxidative stress and an inflammatory attack, and protecting mitochondrial function. Furthermore, we hypothesized that GOPs may promote mitochondrial biosynthesis by upregulating PGC-1α expression. Similarly, GOPs positively regulated cell stability in PC-12; notably, the protective effect of GOPs on PC-12 mainly occurred through the inhibition of autophagic cell death of neuronal cells, while the protective effect on mitochondria was weak. In conclusion, it is evident that GOPs demonstrate potential beneficial effects in maintaining cell fate stability, thereby potentially contributing to an enhanced health span and overall well-being.
Collapse
Affiliation(s)
- Na Zhu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
- College of Public Health, Inner Mongolia Medical University, Hohhot 010059, China
| | - Yong Li
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Meihong Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| |
Collapse
|
166
|
Zheng G, Li C, Chen X, Deng Z, Xie T, Huo Z, Wei X, Huang Y, Zeng X, Luo Y, Bai J. HDAC9 inhibition reduces skeletal muscle atrophy and enhances regeneration in mice with cigarette smoke-induced COPD. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167023. [PMID: 38218381 DOI: 10.1016/j.bbadis.2024.167023] [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: 06/10/2023] [Revised: 01/07/2024] [Accepted: 01/08/2024] [Indexed: 01/15/2024]
Abstract
Cigarette smoke (CS) is the major risk factor for chronic obstructive pulmonary disease (COPD), and sarcopenia is one of the significant comorbidities of COPD. However, the pathogenesis of CS-related deficient skeletal muscle regeneration has yet to be clarified. The impact of CS on myoblast differentiation was examined, and then we determined which HDAC influenced the myogenic process and muscle atrophy in vitro and in vivo. Finally, we further investigated the potential mechanisms via RNA sequencing. Long-term CS exposure activated skeletal muscle primary satellite cells (SCs) while inhibiting differentiation, and defective myogenesis was also observed in C2C12 cells treated with CS extract (CSE). The level of HDAC9 changed in vitro and in vivo in CS exposure models as well as COPD patients, as detected by bioinformatics analysis. Our data showed that CSE impaired myogenic capacity and myotube formation in C2C12 cells via HDAC9. Moreover, inhibition of HDAC9 in mice exposed to CS prevented skeletal muscle dysfunction and promoted SC differentiation. The results of RNA-Seq analysis and verification indicated that HDAC9 knockout improved muscle differentiation in CS-exposed mice, probably by acting on the AKT/mTOR pathway and inhibiting the P53/P21 pathway. More importantly, the serum of HDAC9 KO mice exposed to CS alleviated the differentiation impairment of C2C12 cells caused by serum intervention in CS-exposed mice, and this effect was inhibited by LY294002 (an AKT/mTOR pathway inhibitor). These results suggest that HDAC9 plays an essential role in the defective regeneration induced by chronic exposure to CS.
Collapse
Affiliation(s)
- Guixian Zheng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Chao Li
- Department of Respiratory Medicine, Hunan Provincial People's Hospital, Changsha, Hunan 410219, China
| | - Xiaoli Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Zhaohui Deng
- Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Zhuzhou, Hunan 412000, China
| | - Ting Xie
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Zengyu Huo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Xinyan Wei
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Yanbing Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Xia Zeng
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, China
| | - Yu Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, China
| | - Jing Bai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China.
| |
Collapse
|
167
|
Huang Y, Lei L, Zhu J, Zheng J, Li Z, Wang H, Wang J, Zheng Z. Pain behavior and phenotype in a modified anterior lumbar disc puncture mouse model. JOR Spine 2024; 7:e1284. [PMID: 38249720 PMCID: PMC10797215 DOI: 10.1002/jsp2.1284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 09/03/2023] [Accepted: 09/06/2023] [Indexed: 01/23/2024] Open
Abstract
Background An experimental study was performed to improve the anterior approach model of intervertebral disc degeneration (IVDD). Objective The aims of this study were to investigate the anterior approach model of IVDD for the cause of death, phenotypes, and underlying mechanisms of low back pain in mice. Method In this study, we conducted an anterior puncture procedure on a cohort of 300 C57BL/6J mice that were 8 weeks old. Our investigation focused on exploring the causes of death in the study population (n = 300) and assessing the time-course changes in various parameters, including radiographical, histological, immunofluorescence, and immunohistochemistry analyses (n = 10). Additionally, we conducted behavioral assessments on a subset of the animals (n = 30). Results Transverse vertebral artery rupture is a major factor in surgical death. Radiographical analyses showed that the hydration of the nucleus pulposus began to decrease at 2 weeks after puncture and obviously disappeared over 4 weeks. 3D-CT showed that disc height was significantly decreased at 4 weeks. Osteophyte at the anterior vertebral rims was observed at 2 weeks after the puncture. As the time course increased, histological analyses showed progressive disruption of the destruction of the extracellular matrix and increased secretion of inflammatory cytokines and apoptosis. Behavioral signs of low back pain were increased between the puncture and sham groups at 4 weeks. Conclusion The improvement of anterior intervertebral disc approach model in mice will be useful to investigate underlying mechanisms and potential therapeutic strategies for behavior and phenotypes. Furthermore, the application of vibrational pre-treatment can be used to increase the sensitivity of axial back pain in the model, thereby providing researchers with a reliable method for measuring this critical phenotype.
Collapse
Affiliation(s)
- Yuming Huang
- Department of Spine SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
- Sun Yan Sen UniversityPain Research CenterGuangzhouChina
- Guangdong Province Key Laboratory of Orthopaedics and TraumatologyGuangzhouChina
| | - Linchuan Lei
- Department of Spine SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
- Sun Yan Sen UniversityPain Research CenterGuangzhouChina
- Laboratory of General Surgery, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Jian Zhu
- Department of Spine SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
- Sun Yan Sen UniversityPain Research CenterGuangzhouChina
- Guangdong Province Key Laboratory of Orthopaedics and TraumatologyGuangzhouChina
| | - Jinjian Zheng
- Department of Spine SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
- Sun Yan Sen UniversityPain Research CenterGuangzhouChina
- Guangdong Province Key Laboratory of Orthopaedics and TraumatologyGuangzhouChina
| | - Zemin Li
- Department of Spine SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
- Sun Yan Sen UniversityPain Research CenterGuangzhouChina
- Guangdong Province Key Laboratory of Orthopaedics and TraumatologyGuangzhouChina
| | - Hua Wang
- Department of Spine SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
- Sun Yan Sen UniversityPain Research CenterGuangzhouChina
- Guangdong Province Key Laboratory of Orthopaedics and TraumatologyGuangzhouChina
| | - Jianru Wang
- Department of Spine SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
- Sun Yan Sen UniversityPain Research CenterGuangzhouChina
| | - Zhaomin Zheng
- Department of Spine SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
- Sun Yan Sen UniversityPain Research CenterGuangzhouChina
- Guangdong Province Key Laboratory of Orthopaedics and TraumatologyGuangzhouChina
| |
Collapse
|
168
|
Escriche-Navarro B, Garrido E, Sancenón F, García-Fernández A, Martínez-Máñez R. A navitoclax-loaded nanodevice targeting matrix metalloproteinase-3 for the selective elimination of senescent cells. Acta Biomater 2024; 176:405-416. [PMID: 38185231 DOI: 10.1016/j.actbio.2024.01.002] [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: 10/10/2023] [Revised: 12/18/2023] [Accepted: 01/01/2024] [Indexed: 01/09/2024]
Abstract
Cellular senescence is implicated in the occurrence and progression of multiple age-related disorders. In this context, the selective elimination of senescent cells, senolysis, has emerged as an effective therapeutic strategy. However, the heterogeneous senescent phenotype hinders the discovery of a universal and robust senescence biomarker that limits the effective of senolytic with off-target toxic effects. Therefore, the development of more selective strategies represents a promising approach to increase the specificity of senolytic therapy. In this study, we have developed an innovative nanodevice for the selective elimination of senescent cells (SCs) based on the specific enzymatic activity of the senescent secretome. The results revealed that when senescence is induced in proliferating WI-38 by ionizing radiation (IR), the cells secrete high levels of matrix metalloproteinase-3 (MMP-3). Based on this result, mesoporous silica nanoparticles (MSNs) were loaded with the senolytic navitoclax (Nav) and coated with a specific peptide which is substrate of MMP-3 (NPs(Nav)@MMP-3). Studies in cells confirmed the preferential release of cargo in IR-induced senescent cells compared to proliferating cells, depending on MMP-3 levels. Moreover, treatment with NPs(Nav)@MMP-3 induced a selective decrease in the viability of SCs as well as a protective effect on non-proliferating cells. These results demonstrate the potential use of NPs to develop enhanced senolytic therapies based on specific enzymatic activity in the senescent microenvironment, with potential clinical relevance. STATEMENT OF SIGNIFICANCE: The common β-galactosidase activity has been exploited to develop nanoparticles for the selective elimination of senescent cells. However, the identification of new senescent biomarkers is a key factor for the development of improved strategies. In this scenario, we report for the first time the development of NPs targeting senescent cells based on specific enzymatic activity of the senescent secretome. We report a navitoclax-loaded nanodevice responsive to the matrix metalloproteinase-3 (MMP-3) associated with the senescent phenotype. Our nanosystem achieves the selective release of navitoclax in an MMP-3-dependent manner while limiting off-target effects on non-senescent cells. This opens the possibility of using nanoparticles able to detect an altered senescent environment and selectively release its content, thus enhancing the efficacy of senolytic therapies.
Collapse
Affiliation(s)
- Blanca Escriche-Navarro
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM) Universitat Politècnica de València, Universitat de València, Camino de Vera, s/n. 46022, Valencia, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029 Madrid, Spain; Unidad Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València, IIS La Fe. Av. Fernando Abril Martorell, 106 Torre A 7ª planta, 46026, Valencia, Spain
| | - Eva Garrido
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM) Universitat Politècnica de València, Universitat de València, Camino de Vera, s/n. 46022, Valencia, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029 Madrid, Spain
| | - Félix Sancenón
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM) Universitat Politècnica de València, Universitat de València, Camino de Vera, s/n. 46022, Valencia, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, C/ Eduardo Primo Yúfera 3, 46012, Valencia, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029 Madrid, Spain; Unidad Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València, IIS La Fe. Av. Fernando Abril Martorell, 106 Torre A 7ª planta, 46026, Valencia, Spain
| | - Alba García-Fernández
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM) Universitat Politècnica de València, Universitat de València, Camino de Vera, s/n. 46022, Valencia, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, C/ Eduardo Primo Yúfera 3, 46012, Valencia, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029 Madrid, Spain.
| | - Ramón Martínez-Máñez
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM) Universitat Politècnica de València, Universitat de València, Camino de Vera, s/n. 46022, Valencia, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, C/ Eduardo Primo Yúfera 3, 46012, Valencia, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029 Madrid, Spain; Unidad Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València, IIS La Fe. Av. Fernando Abril Martorell, 106 Torre A 7ª planta, 46026, Valencia, Spain.
| |
Collapse
|
169
|
Chen Z, Zhou J, Wu Y, Chen F, Li J, Tao L, Tian Y, Wang H, Li J, Li Z, He W, Zhang K, Wang H. METTL3 promotes cellular senescence of colorectal cancer via modulation of CDKN2B transcription and mRNA stability. Oncogene 2024; 43:976-991. [PMID: 38361047 DOI: 10.1038/s41388-024-02956-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 01/19/2024] [Accepted: 01/25/2024] [Indexed: 02/17/2024]
Abstract
Cellular senescence plays a critical role in cancer development, but the underlying mechanisms remain poorly understood. Our recent study uncovered that replicative senescent colorectal cancer (CRC) cells exhibit increased levels of mRNA N6-methyladenosine (m6A) and methyltransferase METTL3. Knockdown of METTL3 can restore the senescence-associated secretory phenotype (SASP) of CRC cells. Our findings, which were confirmed by m6A-sequencing and functional studies, demonstrate that the cyclin-dependent kinase inhibitor 2B (CDKN2B, encoding p15INK4B) is a mediator of METTL3-regulated CRC senescence. Specifically, m6A modification at position A413 in the coding sequence (CDS) of CDKN2B positively regulates its mRNA stability by recruiting IGF2BP3 and preventing binding with the CCR4-NOT complex. Moreover, increased METTL3 methylates and stabilizes the mRNA of E2F1, which binds to the -208 to -198 regions of the CDKN2B promoter to facilitate transcription. Inhibition of METTL3 or specifically targeting CDKN2B methylation can suppress CRC senescence. Finally, the METTL3/CDKN2B axis-induced senescence can facilitate M2 macrophage polarization and is correlated with aging and CRC progression. The involvement of METTL3/CDKN2B in cell senescence provides a new potential therapeutic target for CRC treatment and expands our understanding of mRNA methylation's role in cellular senescence.
Collapse
Affiliation(s)
- Zhuojia Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Jiawang Zhou
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - You Wu
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Feng Chen
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Jianing Li
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Lijun Tao
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yifan Tian
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Haoran Wang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Jiexin Li
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Zigang Li
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, 518067, China
| | - Weiling He
- Department of Gastrointestinal Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, Fujian, China.
| | - Kun Zhang
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu Seventh People's Hospital, Affiliated Cancer Hospital of Chengdu Medical College, School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, 610500, China.
| | - Hongsheng Wang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| |
Collapse
|
170
|
Muwanigwa MN, Modamio-Chamarro J, Antony PMA, Gomez-Giro G, Krüger R, Bolognin S, Schwamborn JC. Alpha-synuclein pathology is associated with astrocyte senescence in a midbrain organoid model of familial Parkinson's disease. Mol Cell Neurosci 2024; 128:103919. [PMID: 38307302 DOI: 10.1016/j.mcn.2024.103919] [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: 08/30/2023] [Revised: 01/26/2024] [Accepted: 01/27/2024] [Indexed: 02/04/2024] Open
Abstract
Parkinson's disease (PD) is a complex, progressive neurodegenerative disease characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta in the midbrain. Despite extensive research efforts, the molecular and cellular changes that precede neurodegeneration in PD are poorly understood. To address this, here we describe the use of patient specific human midbrain organoids harboring the SNCA triplication to investigate mechanisms underlying dopaminergic degeneration. Our midbrain organoid model recapitulates key pathological hallmarks of PD, including the aggregation of α-synuclein and the progressive loss of dopaminergic neurons. We found that these pathological hallmarks are associated with an increase in senescence associated cellular phenotypes in astrocytes including nuclear lamina defects, the presence of senescence associated heterochromatin foci, and the upregulation of cell cycle arrest genes. These results suggest a role of pathological α-synuclein in inducing astrosenescence which may, in turn, increase the vulnerability of dopaminergic neurons to degeneration.
Collapse
Affiliation(s)
- Mudiwa N Muwanigwa
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Jennifer Modamio-Chamarro
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Paul M A Antony
- Bioimaging Platform, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Gemma Gomez-Giro
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Rejko Krüger
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Silvia Bolognin
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Jens C Schwamborn
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4367 Belvaux, Luxembourg.
| |
Collapse
|
171
|
Guan L, Eisenmenger A, Crasta KC, Sandalova E, Maier AB. Therapeutic effect of dietary ingredients on cellular senescence in animals and humans: A systematic review. Ageing Res Rev 2024; 95:102238. [PMID: 38382678 DOI: 10.1016/j.arr.2024.102238] [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: 10/12/2023] [Revised: 01/12/2024] [Accepted: 02/16/2024] [Indexed: 02/23/2024]
Abstract
BACKGROUND Cellular senescence has been regarded as a therapeutic target for ageing and age-related diseases. Several senotherapeutic agents have been proposed, including compounds derived from natural products which hold the translational potential to promote healthy ageing. This systematic review examined the association of dietary ingredients with cellular senescence in animals and humans, with an intent to identify dietary ingredients with senotherapeutic potential. METHODS This systematic review was registered at PROSPERO International prospective register of systematic reviews (Reg #: CRD42022338885). The databases PubMed and Embase were systematically searched for key terms related to cellular senescence, senescence markers, diets, nutrients and bioactive compounds. Intervention and observational studies on human and animals investigating the effects of dietary ingredients via oral administration on cellular senescence load were included. The SYRCLE's risk of bias tool and Cochrane risk of bias tool v2.0 were used to assess the risk of bias for animal and human studies respectively. RESULTS Out of 5707 identified articles, 83 articles consisting of 78 animal studies and 5 human studies aimed to reduce cellular senescence load using dietary ingredients. In animal studies, the most-frequently used senescence model was normative ageing (26 studies), followed by D-galactose-induced models (17 studies). Resveratrol (8 studies), vitamin E (4 studies) and soy protein isolate (3 studies) showed positive effects on reducing the level of senescence markers such as p53, p21, p16 and senescence-associated ß-galactosidase in various tissues of physiological systems. In three out of five human studies, ginsenoside Rg1 had no positive effect on reducing senescence in muscle tissues after exercise. The risk of bias for both animal and human studies was largely unclear. CONCLUSION Resveratrol, vitamin E and soy protein isolate are promising senotherapeutics studied in animal models. Studies testing dietary ingredients with senotherapeutic potential in humans are limited and translation is highly warranted.
Collapse
Affiliation(s)
- Lihuan Guan
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Centre for Healthy Longevity, @AgeSingapore, National University Health System, Singapore
| | - Anna Eisenmenger
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Centre for Healthy Longevity, @AgeSingapore, National University Health System, Singapore
| | - Karen C Crasta
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Centre for Healthy Longevity, @AgeSingapore, National University Health System, Singapore; Department of Physiology, National University of Singapore, Singapore; NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Agency for Science, Technology & Research (A⁎STAR), Institute of Molecular and Cell Biology (IMCB), Singapore
| | - Elena Sandalova
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Centre for Healthy Longevity, @AgeSingapore, National University Health System, Singapore
| | - Andrea B Maier
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Centre for Healthy Longevity, @AgeSingapore, National University Health System, Singapore; Department of Human Movement Sciences, @AgeAmsterdam, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, the Netherlands.
| |
Collapse
|
172
|
Yao L, Xu Z, Davies DE, Jones MG, Wang Y. Dysregulated bidirectional epithelial-mesenchymal crosstalk: a core determinant of lung fibrosis progression. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2024; 2:27-33. [PMID: 38558961 PMCID: PMC7615773 DOI: 10.1016/j.pccm.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Progressive lung fibrosis is characterised by dysregulated extracellular matrix (ECM) homeostasis. Understanding of disease pathogenesis remains limited and has prevented the development of effective treatments. While an abnormal wound healing response is strongly implicated in lung fibrosis initiation, factors that determine why fibrosis progresses rather than regular tissue repair occurs are not fully explained. Within human lung fibrosis there is evidence of altered epithelial and mesenchymal lung populations as well as cells undergoing epithelial-mesenchymal transition (EMT), a dynamic and reversible biological process by which epithelial cells lose their cell polarity and down-regulate cadherin-mediated cell-cell adhesion to gain migratory properties. This review will focus upon the role of EMT and dysregulated epithelial-mesenchymal crosstalk in progressive lung fibrosis.
Collapse
Affiliation(s)
- Liudi Yao
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Zijian Xu
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Donna E. Davies
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton SO16 6YD, UK
| | - Mark G. Jones
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton SO16 6YD, UK
| | - Yihua Wang
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton SO16 6YD, UK
| |
Collapse
|
173
|
Iske J, Roesel MJ, Martin F, Schroeter A, Matsunaga T, Maenosono R, Tripathi U, Xiao Y, Nian Y, Caldarone BJ, Vondran FWR, Sage PT, Azuma H, Abdi R, Elkhal A, Pirtskhalava T, Tchkonia T, Kirkland JL, Zhou H, Tullius SG. Transplanting old organs promotes senescence in young recipients. Am J Transplant 2024; 24:391-405. [PMID: 37913871 PMCID: PMC10922683 DOI: 10.1016/j.ajt.2023.10.013] [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: 01/01/2023] [Revised: 10/05/2023] [Accepted: 10/16/2023] [Indexed: 11/03/2023]
Abstract
In clinical organ transplantation, donor and recipient ages may differ substantially. Old donor organs accumulate senescent cells that have the capacity to induce senescence in naïve cells. We hypothesized that the engraftment of old organs may induce senescence in younger recipients, promoting age-related pathologies. When performing isogeneic cardiac transplants between age-mismatched C57BL/6 old donor (18 months) mice and young and middle-aged C57BL/6 (3- or 12- month-old) recipients , we observed augmented frequencies of senescent cells in draining lymph nodes, adipose tissue, livers, and hindlimb muscles 30 days after transplantation. These observations went along with compromised physical performance and impaired spatial learning and memory abilities. Systemic levels of the senescence-associated secretory phenotype factors, including mitochondrial DNA (mt-DNA), were elevated in recipients. Of mechanistic relevance, injections of mt-DNA phenocopied effects of age-mismatched organ transplantation on accelerating aging. Single treatment of old donor animals with senolytics prior to transplantation attenuated mt-DNA release and improved physical capacities in young recipients. Collectively, we show that transplanting older organs induces senescence in transplant recipients, resulting in compromised physical and cognitive capacities. Depleting senescent cells with senolytics, in turn, represents a promising approach to improve outcomes of older organs.
Collapse
Affiliation(s)
- Jasper Iske
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Klinik für Herz-, Thorax-, und Gefäßchirurgie, Deutsches Herzzentrum der Charité, Berlin, Germany; Berlin Institutes of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Maximilian J Roesel
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Klinik für Herz-, Thorax-, und Gefäßchirurgie, Deutsches Herzzentrum der Charité, Berlin, Germany
| | - Friederike Martin
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Surgery, CVK/CCM, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Andreas Schroeter
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Regenerative Medicine and Experimental Surgery, Department of General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany
| | - Tomohisa Matsunaga
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Urology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Ryoichi Maenosono
- Department of Urology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Utkarsh Tripathi
- Department of Physiology and Biochemical Engineering Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
| | - Yao Xiao
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yeqi Nian
- Institute of Transplant Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Barbara J Caldarone
- Mouse Behavior Core, Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - Florian W R Vondran
- Regenerative Medicine and Experimental Surgery, Department of General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany
| | - Peter T Sage
- Transplant Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Haruhito Azuma
- Department of Urology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Reza Abdi
- Transplant Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Abdallah Elkhal
- NAD+ Immunology Laboratory, Huntington Medical Research Institutes, Pasadena, California, USA
| | - Tamar Pirtskhalava
- Department of Physiology and Biochemical Engineering Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
| | - Tamara Tchkonia
- Department of Physiology and Biochemical Engineering Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
| | - James L Kirkland
- Department of Physiology and Biochemical Engineering Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA; Division of General Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Hao Zhou
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Stefan G Tullius
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
174
|
Fan H, Zhang M, Wen J, Wang S, Yuan M, Sun H, Shu L, Yang X, Pu Y, Cai Z. Microglia in brain aging: An overview of recent basic science and clinical research developments. J Biomed Res 2024; 38:122-136. [PMID: 38403286 PMCID: PMC11001587 DOI: 10.7555/jbr.37.20220220] [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: 10/05/2022] [Revised: 12/25/2022] [Accepted: 01/12/2023] [Indexed: 02/27/2024] Open
Abstract
Aging is characterized by progressive degeneration of tissues and organs, and it is positively associated with an increased mortality rate. The brain, as one of the most significantly affected organs, experiences age-related changes, including abnormal neuronal activity, dysfunctional calcium homeostasis, dysregulated mitochondrial function, and increased levels of reactive oxygen species. These changes collectively contribute to cognitive deterioration. Aging is also a key risk factor for neurodegenerative diseases, such as Alzheimer's disease and Parkinson's disease. For many years, neurodegenerative disease investigations have primarily focused on neurons, with less attention given to microglial cells. However, recently, microglial homeostasis has emerged as an important mediator in neurological disease pathogenesis. Here, we provide an overview of brain aging from the perspective of the microglia. In doing so, we present the current knowledge on the correlation between brain aging and the microglia, summarize recent progress of investigations about the microglia in normal aging, Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis, and then discuss the correlation between the senescent microglia and the brain, which will culminate with a presentation of the molecular complexity involved in the microglia in brain aging with suggestions for healthy aging.
Collapse
Affiliation(s)
- Haixia Fan
- Chongqing Medical University, Chongqing 400042, China
- Department of Neurology, Chongqing General Hospital, Chongqing 400013, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing 400013, China
- Department of Neurology, the First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Minheng Zhang
- Department of Gerontology, the First People's Hospital of Jinzhong, Jinzhong, Shanxi 030009, China
| | - Jie Wen
- Chongqing Medical University, Chongqing 400042, China
- Department of Neurology, Chongqing General Hospital, Chongqing 400013, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing 400013, China
| | - Shengyuan Wang
- Chongqing Medical University, Chongqing 400042, China
- Department of Neurology, Chongqing General Hospital, Chongqing 400013, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing 400013, China
| | - Minghao Yuan
- Chongqing Medical University, Chongqing 400042, China
- Department of Neurology, Chongqing General Hospital, Chongqing 400013, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing 400013, China
| | - Houchao Sun
- Department of Neurology, Chongqing General Hospital, Chongqing 400013, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing 400013, China
| | - Liu Shu
- Chongqing Medical University, Chongqing 400042, China
- Department of Neurology, Chongqing General Hospital, Chongqing 400013, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing 400013, China
| | - Xu Yang
- Chongqing Medical University, Chongqing 400042, China
- Department of Neurology, Chongqing General Hospital, Chongqing 400013, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing 400013, China
| | - Yinshuang Pu
- Department of Neurology, Chongqing General Hospital, Chongqing 400013, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing 400013, China
| | - Zhiyou Cai
- Chongqing Medical University, Chongqing 400042, China
- Department of Neurology, Chongqing General Hospital, Chongqing 400013, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing 400013, China
| |
Collapse
|
175
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [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
|
176
|
Hattangady NG, Carter K, Maroni-Rana B, Wang T, Ayers JL, Yu M, Grady WM. Mapping the core senescence phenotype of primary human colon fibroblasts. Aging (Albany NY) 2024; 16:3068-3087. [PMID: 38385965 PMCID: PMC10929841 DOI: 10.18632/aging.205577] [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: 01/15/2024] [Indexed: 02/23/2024]
Abstract
Advanced age is the largest risk factor for many diseases and several types of cancer, including colorectal cancer (CRC). Senescent cells are known to accumulate with age in various tissues, where they can modulate the surrounding tissue microenvironment through their senescence associated secretory phenotype (SASP). Recently, we showed that there is an increased number of senescent cells in the colons of CRC patients and demonstrated that senescent fibroblasts and their SASP create microniches in the colon that are conducive to CRC onset and progression. However, the composition of the SASP is heterogenous and cell-specific, and the precise senescence profile of colon fibroblasts has not been well-defined. To generate a SASP atlas of human colon fibroblasts, we induced senescence in primary human colon fibroblasts using various in vitro methods and assessed the resulting transcriptome. Using RNASequencing and further validation by quantitative RT-PCR and Luminex assays, we define and validate a 'core senescent profile' that might play a significant role in shaping the colon microenvironment. We also performed KEGG analysis and GO analyses to identify key pathways and biological processes that are differentially regulated in colon fibroblast senescence. These studies provide insights into potential driver proteins involved in senescence-associated diseases, like CRC, which may lead to therapies to improve overall health in the elderly and to prevent CRC.
Collapse
Affiliation(s)
- Namita Ganesh Hattangady
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Kelly Carter
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Brett Maroni-Rana
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Ting Wang
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Jessica Lee Ayers
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Ming Yu
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - William M. Grady
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Division of Gastroenterology, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
177
|
Pantelis P, Theocharous G, Veroutis D, Vagena IA, Polyzou A, Thanos DF, Kyrodimos E, Kotsinas A, Evangelou K, Lagopati N, Gorgoulis VG, Kotopoulos N. Pulsed Electromagnetic Fields (PEMFs) Trigger Cell Death and Senescence in Cancer Cells. Int J Mol Sci 2024; 25:2473. [PMID: 38473720 DOI: 10.3390/ijms25052473] [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: 01/20/2024] [Revised: 02/10/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
The currently available anti-cancer therapies, such as gamma-radiation and chemotherapeutic agents, induce cell death and cellular senescence not only in cancer cells but also in the adjacent normal tissue. New anti-tumor approaches focus on limiting the side effects on normal cells. In this frame, the potential anti-tumor properties of Pulsed Electromagnetic Fields (PEMFs) through the irradiation of breast cancer epithelial cells (MCF-7 and MDA-MB-231) and normal fibroblasts (FF95) were investigated. PEMFs had a frequency of 8 Hz, full-square wave type and magnetic flux density of 0.011 T and were applied twice daily for 5 days. The data collected showcase that PEMF application decreases the proliferation rate and viability of breast cancer cells while having the opposite effect on normal fibroblasts. Moreover, PEMF irradiation induces cell death and cellular senescence only in breast cancer cells without any effect in the non-cancerous cells. These findings suggest PEMF irradiation as a novel, non-invasive anti-cancer strategy that, when combined with senolytic drugs, may eliminate both cancer and the remaining senescent cells, while simultaneously avoiding the side effects of the current treatments.
Collapse
Affiliation(s)
- Pavlos Pantelis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens (NKUA), 11527 Athens, Greece
| | - Giorgos Theocharous
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens (NKUA), 11527 Athens, Greece
| | - Dimitris Veroutis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens (NKUA), 11527 Athens, Greece
| | - Ioanna-Aglaia Vagena
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens (NKUA), 11527 Athens, Greece
- Laboratory of Biology, Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens (NKUA), 11527 Athens, Greece
| | - Aikaterini Polyzou
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens (NKUA), 11527 Athens, Greece
| | - Dimitris-Foivos Thanos
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens (NKUA), 11527 Athens, Greece
| | - Efthymios Kyrodimos
- 1st ENT Department, Hippocration Hospital, National and Kapodistrian University of Athens (NKUA), 11527 Athens, Greece
| | - Athanassios Kotsinas
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens (NKUA), 11527 Athens, Greece
| | - Konstantinos Evangelou
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens (NKUA), 11527 Athens, Greece
| | - Nefeli Lagopati
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens (NKUA), 11527 Athens, Greece
- Laboratory of Biology, Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens (NKUA), 11527 Athens, Greece
- Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece
| | - Vassilis G Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens (NKUA), 11527 Athens, Greece
- Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece
- Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
- Faculty Institute for Cancer Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester M20 4GJ, UK
- Faculty of Health and Medical Sciences, University of Surrey, Surrey GU2 7YH, UK
| | - Nicholas Kotopoulos
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens (NKUA), 11527 Athens, Greece
| |
Collapse
|
178
|
Zhu S, Yu Y, Hong Q, Li C, Zhang H, Guo K. Neutrophil Extracellular Traps Upregulate p21 and Suppress Cell Cycle Progression to Impair Endothelial Regeneration after Inflammatory Lung Injury. J Clin Med 2024; 13:1204. [PMID: 38592032 PMCID: PMC10931969 DOI: 10.3390/jcm13051204] [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: 12/08/2023] [Revised: 01/23/2024] [Accepted: 02/10/2024] [Indexed: 04/10/2024] Open
Abstract
Background: Sepsis is a major cause of ICU admissions, with high mortality and morbidity. The lungs are particularly vulnerable to infection and injury, and restoration of vascular endothelial homeostasis after injury is a crucial determinant of outcome. Neutrophil extracellular trap (NET) release strongly correlates with the severity of lung tissue damage. However, little is known about whether NETs affect endothelial cell (EC) regeneration and repair. Methods: Eight- to ten-week-old male C57BL/6 mice were injected intraperitoneally with a sublethal dose of LPS to induce acute lung inflammatory injury or with PBS as a control. Blood samples and lung tissues were collected to detect NET formation and lung endothelial cell proliferation. Human umbilical vein endothelial cells (HUVECs) were used to determine the role of NETs in cell cycle progression in vitro. Results: Increased NET formation and impaired endothelial cell proliferation were observed in mice with inflammatory lung injury following septic endotoxemia. Degradation of NETs with DNase I attenuated lung inflammation and facilitated endothelial regeneration. Mechanistically, NETs induced p21 upregulation and cell cycle stasis to impair endothelial repair. Conclusions: Our findings suggest that NET formation impairs endothelial regeneration and vascular repair through the induction of p21 and cell cycle arrest during inflammatory lung injury.
Collapse
Affiliation(s)
- Shuainan Zhu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (S.Z.); (Y.Y.); (Q.H.); (C.L.)
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai 210000, China
| | - Ying Yu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (S.Z.); (Y.Y.); (Q.H.); (C.L.)
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai 210000, China
| | - Qianya Hong
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (S.Z.); (Y.Y.); (Q.H.); (C.L.)
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai 210000, China
| | - Chenning Li
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (S.Z.); (Y.Y.); (Q.H.); (C.L.)
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai 210000, China
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (S.Z.); (Y.Y.); (Q.H.); (C.L.)
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai 210000, China
| | - Kefang Guo
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (S.Z.); (Y.Y.); (Q.H.); (C.L.)
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai 210000, China
| |
Collapse
|
179
|
Camacho-Encina M, Booth LK, Redgrave RE, Folaranmi O, Spyridopoulos I, Richardson GD. Cellular Senescence, Mitochondrial Dysfunction, and Their Link to Cardiovascular Disease. Cells 2024; 13:353. [PMID: 38391966 PMCID: PMC10886919 DOI: 10.3390/cells13040353] [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: 01/11/2024] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 02/24/2024] Open
Abstract
Cardiovascular diseases (CVDs), a group of disorders affecting the heart or blood vessels, are the primary cause of death worldwide, with an immense impact on patient quality of life and disability. According to the World Health Organization, CVD takes an estimated 17.9 million lives each year, where more than four out of five CVD deaths are due to heart attacks and strokes. In the decades to come, an increased prevalence of age-related CVD, such as atherosclerosis, coronary artery stenosis, myocardial infarction (MI), valvular heart disease, and heart failure (HF) will contribute to an even greater health and economic burden as the global average life expectancy increases and consequently the world's population continues to age. Considering this, it is important to focus our research efforts on understanding the fundamental mechanisms underlying CVD. In this review, we focus on cellular senescence and mitochondrial dysfunction, which have long been established to contribute to CVD. We also assess the recent advances in targeting mitochondrial dysfunction including energy starvation and oxidative stress, mitochondria dynamics imbalance, cell apoptosis, mitophagy, and senescence with a focus on therapies that influence both and therefore perhaps represent strategies with the most clinical potential, range, and utility.
Collapse
Affiliation(s)
- Maria Camacho-Encina
- Vascular Medicine and Biology Theme, Bioscience Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK; (R.E.R.); (O.F.); (G.D.R.)
| | - Laura K. Booth
- Vascular Medicine and Biology Theme, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK; (L.K.B.); (I.S.)
| | - Rachael E. Redgrave
- Vascular Medicine and Biology Theme, Bioscience Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK; (R.E.R.); (O.F.); (G.D.R.)
| | - Omowumi Folaranmi
- Vascular Medicine and Biology Theme, Bioscience Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK; (R.E.R.); (O.F.); (G.D.R.)
| | - Ioakim Spyridopoulos
- Vascular Medicine and Biology Theme, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK; (L.K.B.); (I.S.)
| | - Gavin D. Richardson
- Vascular Medicine and Biology Theme, Bioscience Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK; (R.E.R.); (O.F.); (G.D.R.)
| |
Collapse
|
180
|
Sun DY, Wu WB, Wu JJ, Shi Y, Xu JJ, Ouyang SX, Chi C, Shi Y, Ji QX, Miao JH, Fu JT, Tong J, Zhang PP, Zhang JB, Li ZY, Qu LF, Shen FM, Li DJ, Wang P. Pro-ferroptotic signaling promotes arterial aging via vascular smooth muscle cell senescence. Nat Commun 2024; 15:1429. [PMID: 38365899 PMCID: PMC10873425 DOI: 10.1038/s41467-024-45823-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: 07/15/2023] [Accepted: 02/05/2024] [Indexed: 02/18/2024] Open
Abstract
Senescence of vascular smooth muscle cells (VSMCs) contributes to aging-related cardiovascular diseases by promoting arterial remodelling and stiffness. Ferroptosis is a novel type of regulated cell death associated with lipid oxidation. Here, we show that pro-ferroptosis signaling drives VSMCs senescence to accelerate vascular NAD+ loss, remodelling and aging. Pro-ferroptotic signaling is triggered in senescent VSMCs and arteries of aged mice. Furthermore, the activation of pro-ferroptotic signaling in VSMCs not only induces NAD+ loss and senescence but also promotes the release of a pro-senescent secretome. Pharmacological or genetic inhibition of pro-ferroptosis signaling, ameliorates VSMCs senescence, reduces vascular stiffness and retards the progression of abdominal aortic aneurysm in mice. Mechanistically, we revealed that inhibition of pro-ferroptotic signaling facilitates the nuclear-cytoplasmic shuttling of proliferator-activated receptor-γ and, thereby impeding nuclear receptor coactivator 4-ferrtin complex-centric ferritinophagy. Finally, the activated pro-ferroptotic signaling correlates with arterial stiffness in a human proof-of-concept study. These findings have significant implications for future therapeutic strategies aiming to eliminate vascular ferroptosis in senescence- or aging-associated cardiovascular diseases.
Collapse
Affiliation(s)
- Di-Yang Sun
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University/Naval Medical University, Shanghai, China
- Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Wen-Bin Wu
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Jian-Jin Wu
- Department of Vascular and Endovascular Surgery, Changzheng Hospital, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Yu Shi
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jia-Jun Xu
- Department of Diving and Hyperbaric Medicine, Naval Special Medical Center, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Shen-Xi Ouyang
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chen Chi
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Cardiology, School of Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi Shi
- Shanghai Key Laboratory of Organ Transplantation, Fudan University, Shanghai, China
- Institute of Clinical Science, Zhongshan Hospital Fudan University, Shanghai, China
| | - Qing-Xin Ji
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jin-Hao Miao
- Department of Orthopedic Surgery/Spine Center, Changzheng Hospital Affiliated Hospital of Naval Medical University/Second Military Medical University, Shanghai, China
| | - Jiang-Tao Fu
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Jie Tong
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ping-Ping Zhang
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Jia-Bao Zhang
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University/Naval Medical University, Shanghai, China
- The National Demonstration Center for Experimental Pharmaceutical Education, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Zhi-Yong Li
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University/Naval Medical University, Shanghai, China
- The National Demonstration Center for Experimental Pharmaceutical Education, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Le-Feng Qu
- Department of Vascular and Endovascular Surgery, Changzheng Hospital, Naval Medical University/Second Military Medical University, Shanghai, China.
| | - Fu-Ming Shen
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Dong-Jie Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Pei Wang
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University/Naval Medical University, Shanghai, China.
- The National Demonstration Center for Experimental Pharmaceutical Education, Naval Medical University/Second Military Medical University, Shanghai, China.
| |
Collapse
|
181
|
Nègre-Salvayre A, Salvayre R. Reactive Carbonyl Species and Protein Lipoxidation in Atherogenesis. Antioxidants (Basel) 2024; 13:232. [PMID: 38397830 PMCID: PMC10886358 DOI: 10.3390/antiox13020232] [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: 12/29/2023] [Revised: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Atherosclerosis is a multifactorial disease of medium and large arteries, characterized by the presence of lipid-rich plaques lining the intima over time. It is the main cause of cardiovascular diseases and death worldwide. Redox imbalance and lipid peroxidation could play key roles in atherosclerosis by promoting a bundle of responses, including endothelial activation, inflammation, and foam cell formation. The oxidation of polyunsaturated fatty acids generates various lipid oxidation products such as reactive carbonyl species (RCS), including 4-hydroxy alkenals, malondialdehyde, and acrolein. RCS covalently bind to nucleophilic groups of nucleic acids, phospholipids, and proteins, modifying their structure and activity and leading to their progressive dysfunction. Protein lipoxidation is the non-enzymatic post-translational modification of proteins by RCS. Low-density lipoprotein (LDL) oxidation and apolipoprotein B (apoB) modification by RCS play a major role in foam cell formation. Moreover, oxidized LDLs are a source of RCS, which form adducts on a huge number of proteins, depending on oxidative stress intensity, the nature of targets, and the availability of detoxifying systems. Many systems are affected by lipoxidation, including extracellular matrix components, membranes, cytoplasmic and cytoskeletal proteins, transcription factors, and other components. The mechanisms involved in lipoxidation-induced vascular dysfunction are not fully elucidated. In this review, we focus on protein lipoxidation during atherogenesis.
Collapse
Affiliation(s)
- Anne Nègre-Salvayre
- Inserm Unité Mixte de Recherche (UMR), 1297 Toulouse, Centre Hospitalier Universitaire (CHU) Rangueil—BP 84225, 31432 Toulouse CEDEX 4, France;
- Faculty of Medicine, University of Toulouse, 31432 Toulouse, France
| | - Robert Salvayre
- Inserm Unité Mixte de Recherche (UMR), 1297 Toulouse, Centre Hospitalier Universitaire (CHU) Rangueil—BP 84225, 31432 Toulouse CEDEX 4, France;
- Faculty of Medicine, University of Toulouse, 31432 Toulouse, France
| |
Collapse
|
182
|
Zhong H, Chang L, Pei S, Kang Y, Yang L, Wu Y, Chen N, Luo Y, Zhou Y, Xie J, Xia Y. Senescence-related genes analysis in breast cancer reveals the immune microenvironment and implications for immunotherapy. Aging (Albany NY) 2024; 16:3531-3553. [PMID: 38358910 PMCID: PMC10929821 DOI: 10.18632/aging.205544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 01/09/2024] [Indexed: 02/17/2024]
Abstract
Despite the advent of precision therapy for breast cancer (BRCA) treatment, some individuals are still unable to benefit from it and have poor survival prospects as a result of the disease's high heterogeneity. Cell senescence plays a crucial role in the tumorigenesis, progression, and immune regulation of cancer and has a major impact on the tumor microenvironment. To find new treatment strategies, we aimed to investigate the potential significance of cell senescence in BRCA prognosis and immunotherapy. We created a 9-gene senescence-related signature. We evaluated the predictive power and the role of signatures in the immune microenvironment and infiltration. In vitro tests were used to validate the expression and function of the distinctive critical gene ACTC1. Our risk signature allows BRCA patients to receive a Predictive Risk Signature (PRS), which may be used to further categorize a patient's response to immunotherapy. Compared to conventional clinicopathological characteristics, PRS showed strong predictive efficacy and precise survival prediction. Moreover, PRS subgroups were examined for altered pathways, mutational patterns, and possibly useful medicines. Our research offers suggestions for incorporating senescence-based molecular classification into risk assessment and ICI therapy decision-making.
Collapse
Affiliation(s)
- Hua Zhong
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Lijie Chang
- Department of Neonatal Intensive Care Unit, The Second Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Shengbin Pei
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Yakun Kang
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Lili Yang
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Yifan Wu
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Nuo Chen
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Yicheng Luo
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Yixiao Zhou
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Jiaheng Xie
- Department of Burn and Plastic Surgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Yiqin Xia
- Department of Breast Surgery, The First Affiliated Hospital, Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| |
Collapse
|
183
|
He J, Li J, Li Y, Xu Z, Ma M, Chen H, Chen P, Lv L, Shang X, Liu G. Single-cell transcriptomics identifies senescence-associated secretory phenotype (SASP) features of testicular aging in human. Aging (Albany NY) 2024; 16:3350-3362. [PMID: 38349859 PMCID: PMC10929807 DOI: 10.18632/aging.205538] [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: 10/10/2023] [Accepted: 12/29/2023] [Indexed: 02/15/2024]
Abstract
The male reproductive system experiences degradation with age, predominantly impacting the testes. Testicular aging can result in failure to produce physiological testosterone levels, normal sperm concentrations, or both. However, we cannot predict the onset of testicular aging in advance. Using single-cell RNA sequencing (scRNA-seq) from Gene Expression Omnibus (GEO) database, we conducted cell-cell communication network of human testis between older and young group, indicating Leydig cells' potential role in spermatogenesis microenvironment of aging testis. And we depicted the senescence-Associated Secretory Phenotype (SASP) features of aging testis by identifying differentially expressed senescence-associated secretory phenotype (SASP)-related genes between two group. Notably, IGFBP7 mainly expressed in Leydig cells of those differentially expressed SASP-related genes in aging testis. Furthermore, IGFBP7 protein located in the interstitial compartment of older mice confirmed by immunofluorescence and highly expressed in both human seminal plasma and mouse testis in the older group confirmed through Western blot. Together, our findings suggest that IGFBP7 may be a new biomarker of testicular aging.
Collapse
Affiliation(s)
- Junxian He
- Reproductive Medicine Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China
- Guangdong Engineering Technology Research Center of Fertility Preservation, Guangzhou 510655, China
| | - Jindong Li
- Department of Andrology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing 210002, China
- Department of Urology, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou 570100, China
| | - Yanqing Li
- Reproductive Medicine Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China
- Guangdong Engineering Technology Research Center of Fertility Preservation, Guangzhou 510655, China
| | - Zhenhan Xu
- Reproductive Medicine Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China
- Guangdong Engineering Technology Research Center of Fertility Preservation, Guangzhou 510655, China
| | - Menghui Ma
- Reproductive Medicine Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China
- Guangdong Engineering Technology Research Center of Fertility Preservation, Guangzhou 510655, China
| | - Haicheng Chen
- Reproductive Medicine Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China
- Guangdong Engineering Technology Research Center of Fertility Preservation, Guangzhou 510655, China
| | - Peigen Chen
- Reproductive Medicine Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China
- Guangdong Engineering Technology Research Center of Fertility Preservation, Guangzhou 510655, China
| | - Linyan Lv
- Reproductive Medicine Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China
- Guangdong Engineering Technology Research Center of Fertility Preservation, Guangzhou 510655, China
| | - Xuejun Shang
- Department of Andrology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing 210002, China
| | - Guihua Liu
- Reproductive Medicine Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China
- Guangdong Engineering Technology Research Center of Fertility Preservation, Guangzhou 510655, China
| |
Collapse
|
184
|
Duran I, Pombo J, Sun B, Gallage S, Kudo H, McHugh D, Bousset L, Barragan Avila JE, Forlano R, Manousou P, Heikenwalder M, Withers DJ, Vernia S, Goldin RD, Gil J. Detection of senescence using machine learning algorithms based on nuclear features. Nat Commun 2024; 15:1041. [PMID: 38310113 PMCID: PMC10838307 DOI: 10.1038/s41467-024-45421-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: 06/29/2023] [Accepted: 01/22/2024] [Indexed: 02/05/2024] Open
Abstract
Cellular senescence is a stress response with broad pathophysiological implications. Senotherapies can induce senescence to treat cancer or eliminate senescent cells to ameliorate ageing and age-related pathologies. However, the success of senotherapies is limited by the lack of reliable ways to identify senescence. Here, we use nuclear morphology features of senescent cells to devise machine-learning classifiers that accurately predict senescence induced by diverse stressors in different cell types and tissues. As a proof-of-principle, we use these senescence classifiers to characterise senolytics and to screen for drugs that selectively induce senescence in cancer cells but not normal cells. Moreover, a tissue senescence score served to assess the efficacy of senolytic drugs and identified senescence in mouse models of liver cancer initiation, ageing, and fibrosis, and in patients with fatty liver disease. Thus, senescence classifiers can help to detect pathophysiological senescence and to discover and validate potential senotherapies.
Collapse
Affiliation(s)
- Imanol Duran
- MRC Laboratory of Medical Sciences (LMS), Du Cane Road, London, W12 0NN, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Joaquim Pombo
- MRC Laboratory of Medical Sciences (LMS), Du Cane Road, London, W12 0NN, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Bin Sun
- MRC Laboratory of Medical Sciences (LMS), Du Cane Road, London, W12 0NN, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Suchira Gallage
- MRC Laboratory of Medical Sciences (LMS), Du Cane Road, London, W12 0NN, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- M3 Research Center for Malignome, Metabolome and Microbiome, Faculty of Medicine, University of Tuebingen, Otfried-Müller-Straße 37, 72076, Tübingen, Germany
| | - Hiromi Kudo
- Section for Pathology, Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, W2 1NY, UK
| | - Domhnall McHugh
- MRC Laboratory of Medical Sciences (LMS), Du Cane Road, London, W12 0NN, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Laura Bousset
- MRC Laboratory of Medical Sciences (LMS), Du Cane Road, London, W12 0NN, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Jose Efren Barragan Avila
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Roberta Forlano
- Liver Unit, Section of Hepatology and Gastroenterology, Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, W2 1NY, UK
| | - Pinelopi Manousou
- Liver Unit, Section of Hepatology and Gastroenterology, Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, W2 1NY, UK
| | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- M3 Research Center for Malignome, Metabolome and Microbiome, Faculty of Medicine, University of Tuebingen, Otfried-Müller-Straße 37, 72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180), Eberhard Karls University, Tübingen, Germany
| | - Dominic J Withers
- MRC Laboratory of Medical Sciences (LMS), Du Cane Road, London, W12 0NN, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Santiago Vernia
- MRC Laboratory of Medical Sciences (LMS), Du Cane Road, London, W12 0NN, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Robert D Goldin
- Section for Pathology, Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, W2 1NY, UK
| | - Jesús Gil
- MRC Laboratory of Medical Sciences (LMS), Du Cane Road, London, W12 0NN, UK.
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
185
|
Calabrò A, Accardi G, Aiello A, Caruso C, Galimberti D, Candore G. Senotherapeutics to Counteract Senescent Cells Are Prominent Topics in the Context of Anti-Ageing Strategies. Int J Mol Sci 2024; 25:1792. [PMID: 38339070 PMCID: PMC10855240 DOI: 10.3390/ijms25031792] [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: 12/31/2023] [Revised: 01/28/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
Cellular senescence is implicated in ageing and associated with a broad spectrum of age-related diseases. Importantly, a cell can initiate the senescence program irrespective of the organism's age. Various stress signals, including those defined as ageing hallmarks and alterations leading to cancer development, oncogene activation, or loss of cancer-suppressive functions, can trigger cellular senescence. The primary outcome of these alterations is the activation of nuclear factor (NF)-κB, thereby inducing the senescence-associated secretory phenotype (SASP). Proinflammatory cytokines and chemokines, components of this phenotype, contribute to chronic systemic sterile inflammation, commonly referred to as inflamm-ageing. This inflammation is linked to age-related diseases (ARDs), frailty, and increased mortality in older individuals. Additionally, senescent cells (SCs) accumulate in multiple tissues with age and are believed to underlie the organism functional decline, as demonstrated by models. An escalating effort has been dedicated to identify senotherapeutics that selectively target SCs by inducing apoptosis; these drugs are termed senolytics. Concurrently, small molecules that suppress senescent phenotypes without causing cell death are known as senomorphics. Both natural and synthetic senotherapeutics, along with immunotherapies employing immune cell-mediated clearance of SCs, currently represent the most promising strategies to combat ageing and ARDs. Indeed, it is fascinating to observe that information regarding the immune reaction to SCs indicates that regulation by specific lymphocyte subsets, elevated in the oldest centenarians, plays a role in attaining extreme longevity. Regardless, the application of methods already utilized in cancer treatment, such as CAR cells and monoclonal antibodies, broadens the spectrum of potential approaches to be utilized.
Collapse
Affiliation(s)
- Anna Calabrò
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90134 Palermo, Italy; (A.C.); (G.A.); (A.A.); (G.C.)
| | - Giulia Accardi
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90134 Palermo, Italy; (A.C.); (G.A.); (A.A.); (G.C.)
| | - Anna Aiello
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90134 Palermo, Italy; (A.C.); (G.A.); (A.A.); (G.C.)
| | - Calogero Caruso
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90134 Palermo, Italy; (A.C.); (G.A.); (A.A.); (G.C.)
- Italian Association of Anti-Ageing Physicians, 20133 Milan, Italy;
| | | | - Giuseppina Candore
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90134 Palermo, Italy; (A.C.); (G.A.); (A.A.); (G.C.)
| |
Collapse
|
186
|
Xie Z, Liu C, Sun C, Lu Y, Wu S, Liu Y, Wang Q, Wan Y, Wang Y, Yu M, Meng L, Deng J, Zhang W, Wang Z, Yang C, Yuan Y, Xie Z. A novel biomarker of fibrofatty replacement in dystrophinopathies identified by integrating transcriptome, magnetic resonance imaging, and pathology data. J Cachexia Sarcopenia Muscle 2024; 15:98-111. [PMID: 38146684 PMCID: PMC10834313 DOI: 10.1002/jcsm.13410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/23/2023] [Accepted: 10/03/2023] [Indexed: 12/27/2023] Open
Abstract
BACKGROUND We aimed to analyse genome-wide transcriptome differences between Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD) patients and identify biomarkers that correlate well with muscle magnetic resonance imaging (MRI) and histological fibrofatty replacement in both patients, which have not been reported. METHODS One hundred and one male patients with dystrophinopathies (55 DMD and 46 BMD) were enrolled. Muscle-derived genome-wide RNA-sequencing was performed in 31 DMD patients, 29 BMD patients, and 11 normal controls. Fibrofatty replacement was scored on muscle MRI and histological levels in all patients. A unique pipeline, single-sample gene set enrichment analysis combined with Spearman's rank correlations (ssGSEA-Cor) was developed to identify the most correlated gene signature for fibrofatty replacement. Quantitative real-time PCR (qRT-PCR) analysis, western blot analysis, and single-nucleus RNA-sequencing (snRNA-seq) were performed in the remaining patients to validate the most correlated gene signature. RESULTS Comparative transcriptomic analysis revealed that 31 DMD muscles were characterized by a significant increase of inflammation/immune response and extracellular matrix remodelling compared with 29 BMD muscles (P < 0.05). The ssGSEA-Cor pipeline revealed that the gene set of CDKN2A and CDKN2B was the most correlated gene signature for fibrofatty replacement (histological rs = 0.744, P < 0.001; MRI rs = 0.718, P < 0.001). Muscle qRT-PCR confirmed that CDKN2A mRNA expression in both 15 DMD (median = 25.007, P < 0.001) and 12 BMD (median = 5.654, P < 0.001) patients were significantly higher than that in controls (median = 1.101), while no significant difference in CDKN2B mRNA expression was found among DMD, BMD, and control groups. In the 27 patients, muscle CDKN2A mRNA expression respectively correlated with muscle MRI (rs = 0.883, P < 0.001) and histological fibrofatty replacement (rs = 0.804, P < 0.001) and disease duration (rs = 0.645, P < 0.001) and North Star Ambulatory Assessment total scores (rs = -0.698, P < 0.001). Muscle western blot analysis confirmed that both four DMD (median = 2.958, P < 0.05) and four BMD (median = 1.959, P < 0.01) patients had a significantly higher level of CDKN2A protein expression than controls (median = 1.068). The snRNA-seq analysis of two DMD muscles revealed that CDKN2A was mainly expressed in fibro-adipogenic progenitors, satellite cells, and myoblasts. CONCLUSIONS We identify CDKN2A expression as a novel biomarker of fibrofatty replacement, which might be a new target for antifibrotic therapy in dystrophinopathies.
Collapse
Affiliation(s)
- Zhihao Xie
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth HospitalSichuan UniversityChengduChina
| | - Chang Liu
- Department of NeurologyPeking University First HospitalBeijingChina
| | - Chengyue Sun
- Department of NeurologyPeking University People's HospitalBeijingChina
| | - Yanyu Lu
- Department of NeurologyPeking University First HospitalBeijingChina
| | - Shiyi Wu
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth HospitalSichuan UniversityChengduChina
| | - Yilin Liu
- Department of PathologyPeking Union Medical College HospitalBeijingChina
| | - Qi Wang
- Department of NeurologyPeking University First HospitalBeijingChina
| | - Yalan Wan
- Department of NeurologyPeking University First HospitalBeijingChina
| | - Yikang Wang
- Department of NeurologyPeking University First HospitalBeijingChina
| | - Meng Yu
- Department of NeurologyPeking University First HospitalBeijingChina
| | - Lingchao Meng
- Department of NeurologyPeking University First HospitalBeijingChina
| | - Jianwen Deng
- Department of NeurologyPeking University First HospitalBeijingChina
| | - Wei Zhang
- Department of NeurologyPeking University First HospitalBeijingChina
| | - Zhaoxia Wang
- Department of NeurologyPeking University First HospitalBeijingChina
| | - Chunxia Yang
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth HospitalSichuan UniversityChengduChina
| | - Yun Yuan
- Department of NeurologyPeking University First HospitalBeijingChina
| | - Zhiying Xie
- Department of NeurologyPeking University First HospitalBeijingChina
| |
Collapse
|
187
|
Sarandy MM, Gonçalves RV, Valacchi G. Cutaneous Redox Senescence. Biomedicines 2024; 12:348. [PMID: 38397950 PMCID: PMC10886899 DOI: 10.3390/biomedicines12020348] [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: 12/04/2023] [Revised: 01/24/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
Our current understanding of skin cell senescence involves the role of environmental stressors (UV, O3, cigarette smoke, particulate matter, etc.), lifestyle (diet, exercise, etc.) as well as genetic factors (metabolic changes, hormonal, etc.). The common mechanism of action of these stressors is the disturbance of cellular redox balance characterized by increased free radicals and reactive oxygen species (ROS), and when these overload the intrinsic antioxidant defense system, it can lead to an oxidative stress cellular condition. The main redox mechanisms that activate cellular senescence in the skin involve (1) the oxidative damage of telomeres causing their shortening; (2) the oxidation of proteomes and DNA damage; (3) an a in lysosomal mass through the increased activity of resident enzymes such as senescence-associated β-galactosidase (SA-β-gal) as well as other proteins that are products of lysosomal activity; (4) and the increased expression of SASP, in particular pro-inflammatory cytokines transcriptionally regulated by NF-κB. However, the main targets of ROS on the skin are the proteome (oxi-proteome), followed by telomeres, nucleic acids (DNAs), lipids, proteins, and cytoplasmic organelles. As a result, cell cycle arrest pathways, lipid peroxidation, increased lysosomal content and dysfunctional mitochondria, and SASP synthesis occur. Furthermore, oxidative stress in skin cells increases the activity of p16INK4A and p53 as inhibitors of Rb and CDks, which are important for maintaining the cell cycle. p53 also promotes the inactivation of mTOR-mediated autophagic and apoptotic pathways, leading to senescence. However, these markers alone cannot establish the state of cellular senescence, and multiple analyses are encouraged for confirmation. An updated and more comprehensive approach to investigating skin senescence should include further assays of ox-inflammatory molecular pathways that can consolidate the understanding of cutaneous redox senescence.
Collapse
Affiliation(s)
- Mariáurea Matias Sarandy
- Department of Animal Science, Plants for Human Health Institute, North Carolina State University, North Carolina Research Campus, 600 Laureate Way, Kannapolis, NC 28081, USA
- Department of General Biology, Federal University of Viçosa, Viçosa 36570-900, MG, Brazil
| | - Reggiani Vilela Gonçalves
- Department of General Biology, Federal University of Viçosa, Viçosa 36570-900, MG, Brazil
- Department of Animal Biology, Federal University of Viçosa, Viçosa 36570-900, MG, Brazil
| | - Giuseppe Valacchi
- Department of Animal Science, Plants for Human Health Institute, North Carolina State University, North Carolina Research Campus, 600 Laureate Way, Kannapolis, NC 28081, USA
- Department of Environment and Prevention, University of Ferrara, 44121 Ferrara, Italy
- Department of Food and Nutrition, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
188
|
Zhu Y, Anastasiadis ZP, Espindola Netto JM, Evans T, Tchkonia T, Kirkland JL. Past and Future Directions for Research on Cellular Senescence. Cold Spring Harb Perspect Med 2024; 14:a041205. [PMID: 37734865 PMCID: PMC10835613 DOI: 10.1101/cshperspect.a041205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
Cellular senescence was initially described in the early 1960s by Hayflick and Moorehead. They noted sustained cell-cycle arrest after repeated subculturing of human primary cells. Over half a century later, cellular senescence has become recognized as one of the fundamental pillars of aging. Developing senotherapeutics, interventions that selectively eliminate or target senescent cells, has emerged as a key focus in health research. In this article, we note major milestones in cellular senescence research, discuss current challenges, and point to future directions for this rapidly growing field.
Collapse
Affiliation(s)
- Yi Zhu
- Department of Physiology and Biomedical Engineering, Department of Medicine, Mayo Clinic, Rochester, Minnesota 55905, USA
- Robert and Arlene Kogod Center on Aging, Department of Medicine, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Zacharias P Anastasiadis
- Department of Biochemistry and Molecular Biology, Department of Medicine, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | - Tamara Evans
- Robert and Arlene Kogod Center on Aging, Department of Medicine, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Tamar Tchkonia
- Department of Physiology and Biomedical Engineering, Department of Medicine, Mayo Clinic, Rochester, Minnesota 55905, USA
- Robert and Arlene Kogod Center on Aging, Department of Medicine, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - James L Kirkland
- Department of Physiology and Biomedical Engineering, Department of Medicine, Mayo Clinic, Rochester, Minnesota 55905, USA
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota 55905, USA
| |
Collapse
|
189
|
Liu T, Zhu S, Yang Y, Qin W, Wang Z, Zhao Z, Liu T, Wang X, Duan T, Liu Y, Liu Y, Xia Q, Zhang H, Li N. Oroxylin A ameliorates ultraviolet radiation-induced premature skin aging by regulating oxidative stress via the Sirt1 pathway. Biomed Pharmacother 2024; 171:116110. [PMID: 38198955 DOI: 10.1016/j.biopha.2023.116110] [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: 09/29/2023] [Revised: 12/25/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Skin is susceptible to premature aging in response to ultraviolet (UV) radiation-induced oxidative stress, which can ultimately result in aberrant aging or age-related disorders. Accordingly, strategies that can be adopted to mitigate oxidative stress may contribute to protecting skin from induced aging-related damage, thereby offering promising approaches for the treatment of skin diseases and disorders. In this regard, oroxylin A (OA), a natural flavonoid isolated from certain plants used in traditional Chinese medicine, is considered to have notable antioxidant, anti-inflammatory, and anti-apoptotic properties, and is often used to treat certain inflammatory diseases. To date, however, there has been comparatively little research on the effects of OA with respect skin aging. In this study, we utilized UV radiation-induced mouse and cellular models of aging to assess the efficacy of OA in protecting against skin aging. Subsequently, to elucidate the potential mechanisms underlying the protective effect of OA on skin aging, we performed molecular docking analysis to investigate the involvement of the anti-aging gene Sirt1, which was further confirmed on the basis of Sirt1 gene silencing. We accordingly demonstrated that by promoting an increase in the expression of Sirt1, OA can contribute to suppressing UV-induced skin photo-aging in cells/mice by reducing oxidative stress. Furthermore, we established that by activating Sirt1, OA can also promote the dissociation of Nrf2 from Keap1 and its subsequent nuclear translocation. Collectively, our findings in this study reveal OA to be an effective natural compound that can be administered to delay the aging of skin triggered by UV, both in vivo and in vitro, by binding to Sirt1 to promote the deacetylation and nuclear translocation of Nrf2, thereby contributing to a reduction in oxidative stress. These findings may this provide a therapeutic target for the prevention of skin aging or aging-induced skin diseases.
Collapse
Affiliation(s)
- Tao Liu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, PR China; Laboratory of Pharmacology of TCM Formulae Co-Constructed by the Province-Ministry, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China; National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional chinese Medicine, Tianjin, PR China
| | - Shan Zhu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, PR China; Laboratory of Pharmacology of TCM Formulae Co-Constructed by the Province-Ministry, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China; National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional chinese Medicine, Tianjin, PR China
| | - Yi Yang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, PR China; Laboratory of Pharmacology of TCM Formulae Co-Constructed by the Province-Ministry, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China; National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional chinese Medicine, Tianjin, PR China
| | - Wenxiao Qin
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, PR China; Laboratory of Pharmacology of TCM Formulae Co-Constructed by the Province-Ministry, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China; National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional chinese Medicine, Tianjin, PR China
| | - Zijing Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, PR China; Laboratory of Pharmacology of TCM Formulae Co-Constructed by the Province-Ministry, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China; National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional chinese Medicine, Tianjin, PR China
| | - Zhiyue Zhao
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, PR China; Laboratory of Pharmacology of TCM Formulae Co-Constructed by the Province-Ministry, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China; National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional chinese Medicine, Tianjin, PR China
| | - Tao Liu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, PR China; Laboratory of Pharmacology of TCM Formulae Co-Constructed by the Province-Ministry, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China; National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional chinese Medicine, Tianjin, PR China
| | - Xiang Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, PR China; Laboratory of Pharmacology of TCM Formulae Co-Constructed by the Province-Ministry, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China; National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional chinese Medicine, Tianjin, PR China
| | - Tian Duan
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, PR China; Laboratory of Pharmacology of TCM Formulae Co-Constructed by the Province-Ministry, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China; National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional chinese Medicine, Tianjin, PR China
| | - Yang Liu
- Chinese medical college,Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Yan Liu
- Tianjin Polytechnic University, Tianjin, PR China
| | - Qingmei Xia
- Chinese medical college,Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Han Zhang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, PR China; Laboratory of Pharmacology of TCM Formulae Co-Constructed by the Province-Ministry, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China; National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional chinese Medicine, Tianjin, PR China.
| | - Nan Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, PR China; Laboratory of Pharmacology of TCM Formulae Co-Constructed by the Province-Ministry, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China; National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional chinese Medicine, Tianjin, PR China; Engineering research center of Modern chinese Medicine Discovery and Preparation Technique, Ministry of education, Tianjin University of Traditional chinese Medicine, Tianjin, PR China.
| |
Collapse
|
190
|
Hao X, Zhao B, Towers M, Liao L, Monteiro EL, Xu X, Freeman C, Peng H, Tang HY, Havas A, Kossenkov AV, Berger SL, Adams PD, Speicher DW, Schultz D, Marmorstein R, Zaret KS, Zhang R. TXNRD1 drives the innate immune response in senescent cells with implications for age-associated inflammation. NATURE AGING 2024; 4:185-197. [PMID: 38267705 PMCID: PMC11210448 DOI: 10.1038/s43587-023-00564-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 12/22/2023] [Indexed: 01/26/2024]
Abstract
Sterile inflammation, also known as 'inflammaging', is a hallmark of tissue aging. Cellular senescence contributes to tissue aging, in part, through the secretion of proinflammatory factors collectively known as the senescence-associated secretory phenotype (SASP). The genetic variability of thioredoxin reductase 1 (TXNRD1) is associated with aging and age-associated phenotypes such as late-life survival, activity of daily living and physical performance in old age. TXNRD1's role in regulating tissue aging has been attributed to its enzymatic role in cellular redox regulation. Here, we show that TXNRD1 drives the SASP and inflammaging through the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) innate immune response pathway independently of its enzymatic activity. TXNRD1 localizes to cytoplasmic chromatin fragments and interacts with cGAS in a senescence-status-dependent manner, which is necessary for the SASP. TXNRD1 enhances the enzymatic activity of cGAS. TXNRD1 is required for both the tumor-promoting and immune surveillance functions of senescent cells, which are mediated by the SASP in vivo in mouse models. Treatment of aged mice with a TXNRD1 inhibitor that disrupts its interaction with cGAS, but not with an inhibitor of its enzymatic activity alone, downregulated markers of inflammaging in several tissues. In summary, our results show that TXNRD1 promotes the SASP through the innate immune response, with implications for inflammaging. This suggests that the TXNRD1-cGAS interaction is a relevant target for selectively suppressing inflammaging.
Collapse
Affiliation(s)
- Xue Hao
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bo Zhao
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Martina Towers
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Liping Liao
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Edgar Luzete Monteiro
- Penn Epigenetics Institute, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xin Xu
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christina Freeman
- High-throughput Screening Core, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hongzhuang Peng
- High-throughput Screening Core, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hsin-Yao Tang
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA, USA
| | - Aaron Havas
- Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA, USA
| | - Andrew V Kossenkov
- Gene Expression and Regulation Program, The Wistar Institute, Philadelphia, PA, USA
| | - Shelley L Berger
- Penn Epigenetics Institute, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Peter D Adams
- Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA, USA
| | - David W Speicher
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA, USA
| | - David Schultz
- High-throughput Screening Core, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ronen Marmorstein
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kenneth S Zaret
- Penn Epigenetics Institute, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rugang Zhang
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, USA.
| |
Collapse
|
191
|
Bao Q, Yu X, Qi X. Integrated analysis of single-cell sequencing and weighted co-expression network identifies a novel signature based on cellular senescence-related genes to predict prognosis in glioblastoma. ENVIRONMENTAL TOXICOLOGY 2024; 39:643-656. [PMID: 37565732 DOI: 10.1002/tox.23921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/17/2023] [Accepted: 07/21/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND Glioblastoma (GBM) is a highly aggressive cancer with heavy mortality rates and poor prognosis. Cellular senescence exerts a pivotal influence on the development and progression of various cancers. However, the underlying effect of cellular senescence on the outcomes of patients with GBM remains to be elucidated. METHODS Transcriptome RNA sequencing data with clinical information and single-cell sequencing data of GBM cases were obtained from CGGA, TCGA, and GEO (GSE84465) databases respectively. Single-sample gene set enrichment analysis (ssGSEA) analysis was utilized to calculate the cellular senescence score. WGCNA analysis was employed to ascertain the key gene modules and identify differentially expressed genes (DEGs) associated with the cellular senescence score in GBM. The prognostic senescence-related risk model was developed by least absolute shrinkage and selection operator (LASSO) regression analyses. The immune infiltration level was calculated by microenvironment cell populations counter (MCPcounter), ssGSEA, and xCell algorithms. Potential anti-cancer small molecular compounds of GBM were estimated by "oncoPredict" R package. RESULTS A total of 150 DEGs were selected from the pink module through WGCNA analysis. The risk-scoring model was constructed based on 5 cell senescence-associated genes (CCDC151, DRC1, C2orf73, CCDC13, and WDR63). Patients in low-risk group had a better prognostic value compared to those in high-risk group. The nomogram exhibited excellent predictive performance in assessing the survival outcomes of patients with GBM. Top 30 potential anti-cancer small molecular compounds with higher drug sensitivity scores were predicted. CONCLUSION Cellular senescence-related genes and clusters in GBM have the potential to provide valuable insights in prognosis and guide clinical decisions.
Collapse
Affiliation(s)
- Qingquan Bao
- Department of Neurosurgery, Shaoxing People's Hospital, Shaoxing, China
| | - Xuebin Yu
- Department of Neurosurgery, Shaoxing People's Hospital, Shaoxing, China
| | - Xuchen Qi
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
192
|
Pagliuca S, Ferraro F. Immune-driven clonal cell selection at the intersection among cancer, infections, autoimmunity and senescence. Semin Hematol 2024; 61:22-34. [PMID: 38341340 DOI: 10.1053/j.seminhematol.2024.01.002] [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: 11/25/2023] [Revised: 12/14/2023] [Accepted: 01/02/2024] [Indexed: 02/12/2024]
Abstract
Immune surveillance mechanisms play a crucial role in maintaining lifelong immune homeostasis in response to pathologic stimuli and aberrant cell states. However, their persistence, especially in the context of chronic antigenic exposure, can create a fertile ground for immune evasion. These escaping cell phenotypes, harboring a variety of genomic and transcriptomic aberrances, chiefly in human leukocyte antigen (HLA) and antigen presentation machinery genes, may survive and proliferate, featuring a scenario of clonal cell expansion with immune failure characteristics. While well characterized in solid and, to some extent, hematological malignancies, little is known about their occurrence and significance in other disease contexts. Historical literature highlights the role for escaping HLA-mediated recognition as a strategy adopted by virus to evade from the immune system, hinting at the potential for immune aberrant cell expansion in the context of chronic infections. Additionally, unmasked in idiopathic aplastic anemia as a mechanism able to rescue failing hematopoiesis, HLA clonal escape may operate in autoimmune disorders, particularly in tissues targeted by aberrant immune responses. Furthermore, senescent cell status emerging as immunogenic phenotypes stimulating T cell responses, may act as a bottleneck for the selection of such immune escaping clones, blurring the boundaries between neoplastic transformation, aging and inflammation. Here we provide a fresh overview and perspective on this immune-driven clonal cell expansion, linking pathophysiological features of neoplastic, autoimmune, infectious and senescence processes exposed to immune surveillance.
Collapse
Affiliation(s)
- Simona Pagliuca
- Hematology Department, Nancy University Hospital and UMR7365, IMoPA, University of Lorraine, Vandoeuvre-lès-Nancy, France.
| | - Francesca Ferraro
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO
| |
Collapse
|
193
|
Zhao J, Han Z, Ding L, Wang P, He X, Lin L. The molecular mechanism of aging and the role in neurodegenerative diseases. Heliyon 2024; 10:e24751. [PMID: 38312598 PMCID: PMC10835255 DOI: 10.1016/j.heliyon.2024.e24751] [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: 07/03/2023] [Revised: 12/09/2023] [Accepted: 01/12/2024] [Indexed: 02/06/2024] Open
Abstract
Aging is a complex and inevitable biological process affected by a combination of external environmental and genetic factors. Humans are currently living longer than ever before, accompanied with aging-related alterations such as diminished autophagy, decreased immunological function, mitochondrial malfunction, stem cell failure, accumulation of somatic and mitochondrial DNA mutations, loss of telomere, and altered nutrient metabolism. Aging leads to a decline in body functions and age-related diseases, for example, Alzheimer's disease, which adversely affects human health and longevity. The quality of life of the elderly is greatly diminished by the increase in their life expectancy rather than healthy life expectancy. With the rise in the age of the global population, aging and related diseases have become the focus of attention worldwide. In this review, we discuss several major mechanisms of aging, including DNA damage and repair, free radical oxidation, telomeres and telomerase, mitochondrial damage, inflammation, and their role in neurodegenerative diseases to provide a reference for the prevention of aging and its related diseases.
Collapse
Affiliation(s)
- Juanli Zhao
- Laboratory of Medical Molecular and Cellular Biology, College of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, 430065, China
- Department of Pharmacology, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Zhenjie Han
- Laboratory of Medical Molecular and Cellular Biology, College of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Li Ding
- Department of Pharmacology, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Ping Wang
- Hubei Research Institute of Geriatrics, Collaborative Innovation Center of Hubei Province, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Xiutang He
- Center for Monitoring and Evaluation of Teaching Quality, Jingchu University of Technology, Jingmen, 448000, China
| | - Li Lin
- Laboratory of Medical Molecular and Cellular Biology, College of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, 430065, China
| |
Collapse
|
194
|
Liu ZF, Zhang Y, Liu J, Wang YY, Chen M, Liu EY, Guo JM, Wang YH, Weng ZW, Liu CX, Yu CH, Wang XY. Effect of Traditional Chinese Non-Pharmacological Therapies on Knee Osteoarthritis: A Narrative Review of Clinical Application and Mechanism. Orthop Res Rev 2024; 16:21-33. [PMID: 38292459 PMCID: PMC10826518 DOI: 10.2147/orr.s442025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/16/2024] [Indexed: 02/01/2024] Open
Abstract
Knee osteoarthritis (KOA) stands as a degenerative ailment with a substantial and escalating prevalence. The practice of traditional Chinese non-pharmacological therapy has become a prevalent complementary and adjunctive approach. A mounting body of evidence suggests its efficacy in addressing KOA. Recent investigations have delved into its underlying mechanism, yielding some headway. Consequently, this comprehensive analysis seeks to encapsulate the clinical application and molecular mechanism of traditional Chinese non-pharmacological therapy in KOA treatment. The review reveals that various therapies, such as acupuncture, electroacupuncture, warm needle acupuncture, tuina, and acupotomy, primarily target localized knee components like cartilage, subchondral bone, and synovium. Moreover, their impact extends to the central nervous system and intestinal flora. More perfect experimental design and more comprehensive research remain a promising avenue in the future.
Collapse
Affiliation(s)
- Zhi-Feng Liu
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Yang Zhang
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Jing Liu
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Yu-Yan Wang
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Mo Chen
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Er-Yang Liu
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Jun-Ming Guo
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Yan-Hua Wang
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Zhi-Wen Weng
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Chang-Xin Liu
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Chang-He Yu
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| | - Xi-You Wang
- Tuina and Pain Management Department, Beijing University of Chinese Medicine Affilliated Dongzhimen Hospital, Beijing, People’s Republic of China
| |
Collapse
|
195
|
Bartosz G, Pieńkowska N, Sadowska-Bartosz I. Effect of Selected Antioxidants on the In Vitro Aging of Human Fibroblasts. Int J Mol Sci 2024; 25:1529. [PMID: 38338809 PMCID: PMC10855218 DOI: 10.3390/ijms25031529] [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: 01/02/2024] [Revised: 01/17/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
The modification of the replicative lifespan (RLS) of fibroblasts is of interest both from a knowledge point of view and for the attenuation of skin aging. The effect of six antioxidants at a concentration of 1 μM on the replicative lifespan of human dermal fibroblasts was studied. The nitroxide 4-hydroxy-TEMPO (TEMPOL), ergothioneine, and Trolox extended the replicative lifespan (RLS) (40 ± 1 population doublings (PD)) by 7 ± 2, 4 ± 1, and 3 ± 1 PD and lowered the expression of p21 at late passages. Coumaric acid, curcumin and resveratrol did not affect the RLS . The level of reactive oxygen species (ROS) was decreased or not affected by the antioxidants although TEMPOL and coumaric acid decreased the level of glutathione. Only ergothioneine and resveratrol decreased the level of protein carbonylation. The antioxidants that could prolong the RLS elevated the mitochondrial membrane potential. Protecting the activity of mitochondria seems to be important for maintaining the replicative capacity of fibroblasts.
Collapse
Affiliation(s)
| | | | - Izabela Sadowska-Bartosz
- Laboratory of Analytical Biochemistry, Institute of Food Technology and Nutrition, College of Natural Sciences, Rzeszow University, Zelwerowicza Street 4, 35-601 Rzeszow, Poland; (G.B.); (N.P.)
| |
Collapse
|
196
|
Ge Y, Li M, Bai S, Chen C, Zhang S, Cheng J, Wang X. Doxercalciferol alleviates UVB-induced HaCaT cell senescence and skin photoaging. Int Immunopharmacol 2024; 127:111357. [PMID: 38104366 DOI: 10.1016/j.intimp.2023.111357] [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: 08/19/2023] [Revised: 11/23/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
Prolonged or excessive ultraviolet (UV) exposure can lead to premature skin aging. Doxercalciferol (Dox), an analog of vitamin D2, is chiefly used to treat endocrine diseases, cardiovascular diseases, kidney diseases, etc. To date, research on Dox in alleviating photoaging and UV-induced inflammation is scarce. In this research, we evaluated the function of Dox in ultraviolet radiation B (UVB)-induced photoaging and explored the potential mechanism in human keratinocytes (Hacat) and BALB/c mice. First, we established a stable UVB-induced photoaging cell model. Then, we found that the senescence β-galactosidase (SA-β-Gal) positive rate, senescence-related protein (p16), aging-related genes (p21 and p53), senescence-associated secretory phenotype (SASP), inflammatory driving factors (IL-1β and IL-6) and matrix metalloproteinases (MMPs) (MMP1 and MMP9) were upregulated in HaCaT cells after UVB irradiation. At the same time, the effect of UVB on the back skin of BALB/c mice showed a consistent trend. Dox effectively alleviated the aforementioned changes caused by UVB radiation. Mechanistically, we found that UVB activated mitogen-activated protein kinase (MAPK) and nuclear factor kappa B (NF-κB) signaling pathways, and Dox inhibited UVB-activated NF-κB and MAPK. Furthermore, Dox inhibited UVB-induced skin photoaging and damage in mice. In summary, Dox has been improved to inhibit photoaging, which may help to develop therapies to delay skin photoaging.
Collapse
Affiliation(s)
- Yuchen Ge
- School of Basic Medicine, Dali University, Dali, Yunnan 671000, China
| | - Man Li
- School of Basic Medicine, Dali University, Dali, Yunnan 671000, China
| | - Shirui Bai
- School of Basic Medicine, Dali University, Dali, Yunnan 671000, China
| | - Cui Chen
- School of Basic Medicine, Dali University, Dali, Yunnan 671000, China
| | - Shulin Zhang
- School of Basic Medicine, Dali University, Dali, Yunnan 671000, China
| | - Jiamao Cheng
- School of Basic Medicine, Dali University, Dali, Yunnan 671000, China.
| | - Xiaobo Wang
- School of Basic Medicine, Dali University, Dali, Yunnan 671000, China.
| |
Collapse
|
197
|
Fang YP, Zhao Y, Huang JY, Yang X, Liu Y, Zhang XL. The functional role of cellular senescence during vascular calcification in chronic kidney disease. Front Endocrinol (Lausanne) 2024; 15:1330942. [PMID: 38318291 PMCID: PMC10839002 DOI: 10.3389/fendo.2024.1330942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 01/03/2024] [Indexed: 02/07/2024] Open
Abstract
Vascular calcification (VC) has emerged as a key predictor of cardiovascular events in patients with chronic kidney disease (CKD). In recent years, an expanding body of research has put forth the concept of accelerated vascular aging among CKD patients, highlighting the significance of vascular cells senescence in the process of VC. Within the milieu of uremia, senescent vascular endothelial cells (VECs) release extracellular microvesicles (MV) that promote vascular smooth muscle cells (VSMCs) senescence, thereby triggering the subsequent osteogenic phenotypic switch and ultimately contributing to the VC process. In addition, senescent vascular progenitor or stem cells with diminished ability to differentiate into VECs and VSMCS, compromise the repair of vascular integrity, on the other hand, release a cascade of molecules associated with senescence, collectively known as the senescence-associated secretory phenotype (SASP), perpetuating the senescence phenomenon. Furthermore, SASP triggers the recruitment of monocytes and macrophages, as well as adjacent VECs and VSMCs into a pro-adhesive and pro-inflammatory senescent state. This pro-inflammatory microenvironment niche not only impacts the functionality of immune cells but also influences the differentiation of myeloid immune cells, thereby amplifying the reduced ability to effectively clear senescent cells of senescent macrophages, promoted calcification of VSMCs. The objective of this paper is to provide a comprehensive review of the contribution of vascular cell senescence to the emergence and advancement of VC. Gaining a comprehensive understanding of the involvement of cellular senescence within the vessel wall is pivotal, especially when it comes to its intersection with VC. This knowledge is essential for advancing groundbreaking anti-aging therapies, aiming to effectively mitigate cardiovascular diseases.
Collapse
Affiliation(s)
- Ya-Ping Fang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Yu Zhao
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Jia-Yi Huang
- Department of Clinical Medicine, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Xin Yang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Yan Liu
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Xiao-Liang Zhang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
198
|
Ziegler DV, Czarnecka-Herok J, Vernier M, Scholtes C, Camprubi C, Huna A, Massemin A, Griveau A, Machon C, Guitton J, Rieusset J, Vigneron AM, Giguère V, Martin N, Bernard D. Cholesterol biosynthetic pathway induces cellular senescence through ERRα. NPJ AGING 2024; 10:5. [PMID: 38216569 PMCID: PMC10786911 DOI: 10.1038/s41514-023-00128-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 10/30/2023] [Indexed: 01/14/2024]
Abstract
Cellular senescence is a cell program induced by various stresses that leads to a stable proliferation arrest and to a senescence-associated secretory phenotype. Accumulation of senescent cells during age-related diseases participates in these pathologies and regulates healthy lifespan. Recent evidences point out a global dysregulated intracellular metabolism associated to senescence phenotype. Nonetheless, the functional contribution of metabolic homeostasis in regulating senescence is barely understood. In this work, we describe how the mevalonate pathway, an anabolic pathway leading to the endogenous biosynthesis of poly-isoprenoids, such as cholesterol, acts as a positive regulator of cellular senescence in normal human cells. Mechanistically, this mevalonate pathway-induced senescence is partly mediated by the downstream cholesterol biosynthetic pathway. This pathway promotes the transcriptional activity of ERRα that could lead to dysfunctional mitochondria, ROS production, DNA damage and a p53-dependent senescence. Supporting the relevance of these observations, increase of senescence in liver due to a high-fat diet regimen is abrogated in ERRα knockout mouse. Overall, this work unravels the role of cholesterol biosynthesis or level in the induction of an ERRα-dependent mitochondrial program leading to cellular senescence and related pathological alterations.
Collapse
Affiliation(s)
- Dorian V Ziegler
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France
| | - Joanna Czarnecka-Herok
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France
- Equipe Labellisée la Ligue Contre le Cancer, Lyon, France
| | - Mathieu Vernier
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France
- Equipe Labellisée la Ligue Contre le Cancer, Lyon, France
- Goodman Cancer Research Centre, McGill University, Quebec, Montreal, Canada
| | - Charlotte Scholtes
- Goodman Cancer Research Centre, McGill University, Quebec, Montreal, Canada
| | - Clara Camprubi
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France
| | - Anda Huna
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France
- Equipe Labellisée la Ligue Contre le Cancer, Lyon, France
| | - Amélie Massemin
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France
- Equipe Labellisée la Ligue Contre le Cancer, Lyon, France
| | - Audrey Griveau
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France
| | - Christelle Machon
- Biochemistry and Pharmacology-Toxicology Laboratory, Lyon-Sud Hospital, Hospices Civils de Lyon, F-69495, Pierre Bénite, France
| | - Jérôme Guitton
- Biochemistry and Pharmacology-Toxicology Laboratory, Lyon-Sud Hospital, Hospices Civils de Lyon, F-69495, Pierre Bénite, France
| | | | - Arnaud M Vigneron
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France
| | - Vincent Giguère
- Goodman Cancer Research Centre, McGill University, Quebec, Montreal, Canada
- Departments of Biochemistry, Medicine and Oncology, McGill University, Montreal, Quebec, Montreal, Canada
| | - Nadine Martin
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France.
- Equipe Labellisée la Ligue Contre le Cancer, Lyon, France.
| | - David Bernard
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France.
- Equipe Labellisée la Ligue Contre le Cancer, Lyon, France.
| |
Collapse
|
199
|
Saito A, Omura I, Imaizumi K. CREB3L1/OASIS: cell cycle regulator and tumor suppressor. FEBS J 2024. [PMID: 38215153 DOI: 10.1111/febs.17052] [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: 09/11/2023] [Revised: 11/09/2023] [Accepted: 01/05/2024] [Indexed: 01/14/2024]
Abstract
Cell cycle checkpoints detect DNA errors, eventually arresting the cell cycle to promote DNA repair. Failure of such cell cycle arrest causes aberrant cell proliferation, promoting the pathogenesis of multiple diseases, including cancer. Endoplasmic reticulum (ER) stress transducers activate the unfolded protein response, which not only deals with unfolded proteins in ER lumen but also orchestrates diverse physiological phenomena such as cell differentiation and lipid metabolism. Among ER stress transducers, cyclic AMP-responsive element-binding protein 3-like protein 1 (CREB3L1) [also known as old astrocyte specifically induced substance (OASIS)] is an ER-resident transmembrane transcription factor. This molecule is cleaved by regulated intramembrane proteolysis, followed by activation as a transcription factor. OASIS is preferentially expressed in specific cells, including astrocytes and osteoblasts, to regulate their differentiation. In accordance with its name, OASIS was originally identified as being upregulated in long-term-cultured astrocytes undergoing cell cycle arrest because of replicative stress. In the context of cell cycle regulation, previously unknown physiological roles of OASIS have been discovered. OASIS is activated as a transcription factor in response to DNA damage to induce p21-mediated cell cycle arrest. Although p21 is directly induced by the master regulator of the cell cycle, p53, no crosstalk occurs between p21 induction by OASIS or p53. Here, we summarize previously unknown cell cycle regulation by ER-resident transcription factor OASIS, particularly focusing on commonalities and differences in cell cycle arrest between OASIS and p53. This review also mentions tumorigenesis caused by OASIS dysfunctions, and OASIS's potential as a tumor suppressor and therapeutic target.
Collapse
Affiliation(s)
- Atsushi Saito
- Department of Biochemistry, Institute of Biomedical & Health Sciences, Hiroshima University, Japan
| | - Issei Omura
- Department of Biochemistry, Institute of Biomedical & Health Sciences, Hiroshima University, Japan
| | - Kazunori Imaizumi
- Department of Biochemistry, Institute of Biomedical & Health Sciences, Hiroshima University, Japan
| |
Collapse
|
200
|
Chu JH, Xiong J, Wong CTT, Wang S, Tam DY, García-Fernández A, Martínez-Máñez R, Ng DKP. Detection and Elimination of Senescent Cells with a Self-Assembled Senescence-Associated β-Galactosidase-Activatable Nanophotosensitizer. J Med Chem 2024; 67:234-244. [PMID: 38113190 PMCID: PMC10788907 DOI: 10.1021/acs.jmedchem.3c01306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/21/2023]
Abstract
Senescent cells have become an important therapeutic target for many age-related dysfunctions and diseases. We report herein a novel nanophotosensitizing system that is responsive to the senescence-associated β-galactosidase (β-gal) for selective detection and elimination of these cells. It involves a dimeric zinc(II) phthalocyanine linked to a β-galactose unit via a self-immolative linker. This compound can self-assemble in aqueous media, forming stable nanoscale particles in which the phthalocyanine units are stacked and self-quenched for fluorescence emission and singlet oxygen production. Upon internalization into senescent HeLa cells, these nanoparticles interact with the overproduced senescence-associated β-gal inside the cells to trigger the disassembly process through enzymatic cleavage of the glycosidic bonds, followed by self-immolation to release the photoactive monomeric phthalocyanine units. These senescent cells can then be lit up with fluorescence and eliminated through the photodynamic action upon light irradiation with a half-maximal inhibitory concentration of 0.06 μM.
Collapse
Affiliation(s)
- Jacky
C. H. Chu
- Department
of Chemistry, The Chinese University of
Hong Kong, Shatin, N.T., Hong Kong, China
| | - Junlong Xiong
- Department
of Chemistry, The Chinese University of
Hong Kong, Shatin, N.T., Hong Kong, China
- Department
of Pharmacy, The Affiliated Luohu Hospital
of Shenzhen University, Shenzhen University, Shenzhen 518001, China
| | - Clarence T. T. Wong
- Department
of Chemistry, The Chinese University of
Hong Kong, Shatin, N.T., Hong Kong, China
- Department
of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Shuai Wang
- Department
of Chemistry, The Chinese University of
Hong Kong, Shatin, N.T., Hong Kong, China
| | - Dick Yan Tam
- Department
of Chemistry, The Chinese University of
Hong Kong, Shatin, N.T., Hong Kong, China
| | - Alba García-Fernández
- Instituto
Interuniversitario de Investigación de Reconocimiento, Molecular
y Desarrollo Tecnológico, Universitat
Politècnica de València, Universitat de València, Valencia46022, Spain
- CIBER
de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid 28029, Spain
- Unidad Mixta
UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina,
Centro de Investigación Príncipe Felipe, Universitat Politècnica de València, Valencia46012, Spain
| | - Ramón Martínez-Máñez
- Instituto
Interuniversitario de Investigación de Reconocimiento, Molecular
y Desarrollo Tecnológico, Universitat
Politècnica de València, Universitat de València, Valencia46022, Spain
- CIBER
de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid 28029, Spain
- Unidad Mixta
UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina,
Centro de Investigación Príncipe Felipe, Universitat Politècnica de València, Valencia46012, Spain
- Unidad
Mixta de Investigación en Nanomedicina y Sensores, Instituto
de Investigación Sanitaria La Fe (IIS La Fe), Universitat Politècnica e València, Valencia 46026, Spain
| | - Dennis K. P. Ng
- Department
of Chemistry, The Chinese University of
Hong Kong, Shatin, N.T., Hong Kong, China
| |
Collapse
|