1
|
Ateya NH, Al-Taie SF, Jasim SA, Uthirapathy S, Chaudhary K, Rani P, Kundlas M, Naidu KS, Amer NA, Ahmed JK. Histone Deacetylation in Alzheimer's Diseases (AD); Hope or Hype. Cell Biochem Biophys 2025; 83:1537-1553. [PMID: 39825060 DOI: 10.1007/s12013-025-01670-0] [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] [Accepted: 01/05/2025] [Indexed: 01/20/2025]
Abstract
Histone acetylation is the process by which histone acetyltransferases (HATs) add an acetyl group to the N-terminal lysine residues of histones, resulting in a more open chromatin structure. Histone acetylation tends to increase gene expression more than methylation does. In the central nervous system (CNS), histone acetylation is essential for controlling the expression of genes linked to cognition and learning. Histone deacetylases (HDACs), "writing" enzymes (HATs), and "reading" enzymes with bromodomains that identify and localize to acetylated lysine residues are responsible for maintaining histone acetylation. By giving animals HDAC inhibitors (HDACis), it is possible to intentionally control the ratios of "writer" and "eraser" activity, which will change the acetylation of histones. In addition to making the chromatin more accessible, these histone acetylation alterations re-allocate the targeting of "readers," including the transcriptional co-activators, cAMP response element-binding protein (CBP), and bromodomain-containing protein 4 (Brd4) in the CNS. Conclusive evidence has shown that HDACs slow down the progression of Alzheimer's disease (AD) by reducing the amount of histone acetylation, decreasing the activity of genes linked to memory, supporting cognitive decline and Amyloid beta (Aβ) protein accumulation, influencing aberrant tau phosphorylation, and promoting the emergence of neurofibrillary tangles (NFTs). In this review, we have covered the therapeutic targets and functions of HDACs that might be useful in treating AD.
Collapse
Affiliation(s)
- Nabaa Hisham Ateya
- Biotechnology Department, College of Applied Science, Fallujah University, Al-Fallujah, Iraq
| | - Sarah F Al-Taie
- University of Baghdad, College of Science, Department of Biotechnology, Baghdad, Iraq
| | - Saade Abdalkareem Jasim
- Medical Laboratory Techniques department, College of Health and Medical Technology, University of Al-maarif, Anbar, Ramadi, Iraq.
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University Erbil, Kurdistan Region, Erbil, Iraq
| | - Kamlesh Chaudhary
- Department of Neurology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Pooja Rani
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Mayank Kundlas
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - K Satyam Naidu
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, 531162, India
| | - Nevin Adel Amer
- Nursing Department, College of Applied Medical Sciences, Jouf University, Sakakah, Saudi Arabia
- Medical Surgical Nursing Department, Faculty of Nursing, Menofia University, Shibin el Kom, Saudi Arabia
| | - Jawad Kadhim Ahmed
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| |
Collapse
|
2
|
Sang N, Zhang HH, Zhang MY, Zhang MH, Zhu YQ, Chen H, Sun Y, Cheng MC, Wu GC. Prevalence of frailty and prefrailty in systemic lupus erythematosus: A systematic review and meta-analysis. Semin Arthritis Rheum 2025; 72:152709. [PMID: 40086156 DOI: 10.1016/j.semarthrit.2025.152709] [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: 03/25/2024] [Revised: 03/05/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
OBJECTIVE Frailty is associated with mortality in systemic lupus erythematosus (SLE), but the prevalence is unclear. The aim of this systematic review and meta-analysis was to investigate the prevalence of frailty and prefrailty in SLE patients. METHODS Four databases, namely PubMed, Cochrane, EMBASE, and Web of Science, were systematically searched from their inception to November 2024 to identify studies that fulfilled the predefined a priori inclusion criteria for systematic review and meta-analysis, and that specifically investigated frailty and prefrailty in patients with SLE. The quality assessment of the included studies was conducted according to the Newcastle-Ottawa scale (NOS). RESULTS Fifteen studies were retrieved according to the inclusion criteria, and their data were combined in the eventual review. Data from studies including 46,060 patients with SLE were included. The analysis showed that the pooled prevalence of frailty in patients with SLE was 27 % (95 % CI: 19 % - 36 %), the pooled prevalence of prefrailty was 65 % (95 % CI: 54 % - 76 %). Analysis of subgroups revealed that the prevalence of frailty was 26 % (95 % CI: 14 % to 41 %) when measured by Systemic Lupus International Collaborating Clinics-Frailty Index (SLICC-FI) and 22 % (95 % CI: 18 % to 26 %) when assessed using Fried phenotype (FP). CONCLUSION Both frailty and prefrailty are highly prevalent conditions among patients with SLE. There is an urgent need to better understand and address frailty in this population to enhance patient outcomes and quality of life.
Collapse
Affiliation(s)
- Ni Sang
- School of Nursing, Sun Yat-sen University, Guangzhou, PR China.
| | - Hong-Hui Zhang
- Department of Emergency, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, PR China.
| | - Meng-Yao Zhang
- School of Nursing, Anhui Medical University, 15 Feicui Road, Hefei 230032, Anhui, PR China.
| | - Ming-Hui Zhang
- School of Nursing, Anhui Medical University, 15 Feicui Road, Hefei 230032, Anhui, PR China.
| | - Yan-Qin Zhu
- School of Nursing, Anhui Medical University, 15 Feicui Road, Hefei 230032, Anhui, PR China.
| | - Hui Chen
- School of Nursing, Anhui Medical University, 15 Feicui Road, Hefei 230032, Anhui, PR China.
| | - You Sun
- School of Nursing, Anhui Medical University, 15 Feicui Road, Hefei 230032, Anhui, PR China.
| | - Meng-Cheng Cheng
- School of Nursing, Anhui Medical University, 15 Feicui Road, Hefei 230032, Anhui, PR China.
| | - Guo-Cui Wu
- School of Nursing, Anhui Medical University, 15 Feicui Road, Hefei 230032, Anhui, PR China.
| |
Collapse
|
3
|
Greer EL, Lee SS, Prahlad V. Chromatin and epigenetics in aging biology. Genetics 2025; 230:iyaf055. [PMID: 40202900 DOI: 10.1093/genetics/iyaf055] [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: 06/20/2024] [Accepted: 02/03/2025] [Indexed: 04/11/2025] Open
Abstract
This book chapter will focus on modifications to chromatin itself, how chromatin modifications are regulated, and how these modifications are deciphered by the cell to impact aging. In this chapter, we will review how chromatin modifications change with age, examine how chromatin-modifying enzymes have been shown to regulate aging and healthspan, discuss how some of these epigenetic changes are triggered and how they can regulate the lifespan of the individual and its naïve descendants, and speculate on future directions for the field.
Collapse
Affiliation(s)
- Eric Lieberman Greer
- Department of Pediatrics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
- Department of Genetics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Siu Sylvia Lee
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Veena Prahlad
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| |
Collapse
|
4
|
Leeke BJ, Staffhorst I, Percharde M. Emerging roles for the nucleolus in development and stem cells. Development 2025; 152:dev204696. [PMID: 40366093 DOI: 10.1242/dev.204696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
The nucleolus is a membrane-less subnuclear compartment known for its role in ribosome biogenesis. However, emerging evidence suggests that nucleolar function extends beyond ribosome production and is particularly important during mammalian development. Nucleoli are dynamically reprogrammed post-fertilisation: totipotent early mouse embryos display non-canonical, immature nucleolar precursor bodies, and their remodelling to mature nucleoli is essential for the totipotency-to-pluripotency transition. Mounting evidence also links nucleolar disruption to various pathologies, including embryonic lethality in mouse mutants for nucleolar factors, human developmental disorders and observations of nucleolar changes in disease states. As well as its role in ribogenesis, new findings point to the nucleolus as an essential regulator of genome organisation and heterochromatin formation. This Review summarises the varied roles of nucleoli in development, primarily in mammals, highlighting the importance of nucleolar chromatin for genome regulation, and introduces new techniques for exploring nucleolar function.
Collapse
Affiliation(s)
- Bryony J Leeke
- MRC Laboratory of Medical Sciences, London W12 0HS, UK
- Institute of Clinical Sciences, Imperial College London, London W12 0HS, UK
| | - Imke Staffhorst
- MRC Laboratory of Medical Sciences, London W12 0HS, UK
- Institute of Clinical Sciences, Imperial College London, London W12 0HS, UK
| | - Michelle Percharde
- MRC Laboratory of Medical Sciences, London W12 0HS, UK
- Institute of Clinical Sciences, Imperial College London, London W12 0HS, UK
| |
Collapse
|
5
|
Tay JH, Chew YE, Wang W, Lim ZM, Guan L, Dorajoo R, Kennedy BK, Brooke R, Gordevicius J, Horvath S, Sandalova E, Maier AB. DNAm age differences between infinium methylationEPICv1 vs EPICv2 in buffy coat, PBMC, and saliva samples. Commun Biol 2025; 8:654. [PMID: 40269264 PMCID: PMC12019316 DOI: 10.1038/s42003-025-08021-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/31/2025] [Indexed: 04/25/2025] Open
Abstract
This study aims to evaluate differences between Infinium MethylationEPIC (EPICv1) and Infinium MethylationEPICv2 (EPICv2) arrays in estimating DNAm age with eleven DNAm clocks using buffy coat, peripheral blood mononuclear cell (PBMC), and saliva from 16 healthy middle-aged individuals. DNAm ages were estimated using six principal component-based (PC) clocks (PCHorvath1, PCHorvath2, PCHannum, PCPhenoAge, PCGrimAge, and PCDNAmTL) and five non-PC clocks (DunedinPACE, DNAmFit, YingCausAge, YingAdaptAge, and YingDamAge) across all biological samples. Agreement between arrays was assessed using Spearman correlation, Bland-Altman plots, and Wilcoxon Signed-Rank test. The 16 individuals with median age of 48 [43.5;53.8] years, were predominantly female, Chinese and non-smokers. High correlations (ρ > 0.8) were observed between EPICv1 and EPICv2 except for DunedinPACE, YingDamAge and YingAdaptAge. PC-based clocks showed lower systematic bias (MAPE:0.118-8.98%) compared to non-PC-based clocks (MAPE:5.31-21.2%). Saliva samples demonstrated greatest variability between arrays. EPICv2 introduces systematic biases especially in non-PC-based clocks and between different biological samples.
Collapse
Affiliation(s)
- Jian Hua Tay
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
- NUS Academy for Healthy Longevity, National University of Singapore, 10 Medical Drive, 117597, Singapore, Singapore
| | - Yi Ern Chew
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
- NUS Academy for Healthy Longevity, National University of Singapore, 10 Medical Drive, 117597, Singapore, Singapore
| | - Weilan Wang
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
- NUS Academy for Healthy Longevity, National University of Singapore, 10 Medical Drive, 117597, Singapore, Singapore
| | - Zhi Meng Lim
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
- Centre for Healthy Longevity, National University Health System (NUHS), Singapore, 159964, Singapore
| | - Lihuan Guan
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
- NUS Academy for Healthy Longevity, National University of Singapore, 10 Medical Drive, 117597, Singapore, Singapore
| | - Rajkumar Dorajoo
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, 138672, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Brian K Kennedy
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
- Centre for Healthy Longevity, National University Health System (NUHS), Singapore, 159964, Singapore
| | - Robert Brooke
- Epigenetic Clock Development Foundation, Torrance, CA, USA
| | | | - Steve Horvath
- Epigenetic Clock Development Foundation, Torrance, CA, USA
- Altos Labs, San Diego Institute of Science, San Diego, CA, USA
| | - Elena Sandalova
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Andrea B Maier
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore.
- Department of Human Movement Sciences, @AgeAmsterdam, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, Netherlands.
- NUS Academy for Healthy Longevity, National University of Singapore, 10 Medical Drive, 117597, Singapore, Singapore.
| |
Collapse
|
6
|
Della Valle F, Reddy P, Aguirre Vazquez A, Izpisua Belmonte JC. Reactivation of retrotransposable elements is associated with environmental stress and ageing. Nat Rev Genet 2025:10.1038/s41576-025-00829-y. [PMID: 40175591 DOI: 10.1038/s41576-025-00829-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2025] [Indexed: 04/04/2025]
Abstract
Retrotransposable elements (RTEs) are interspersed repetitive sequences that represent a large portion of eukaryotic genomes. Ancestral expansions of RTEs directly contributed to the shaping of these genomes and to the evolution of different species, particularly mammals. RTE activity is tightly regulated by different epigenetic mechanisms but this control becomes compromised as cells age and RTEs are reactivated. This dysregulation of RTEs leads to perturbation of cell function and organ and organismal homeostasis, which drives ageing and age-related disease. Environmental stress is associated with both ageing-related characteristics and the epigenetic mechanisms that control RTE activity, with accumulating evidence indicating that RTE reactivation mediates the effects of environmental stressors on ageing onset and progression. A better understanding of how RTEs are reactivated and their subsequent biological roles may help the development of therapies against ageing-related phenotypes and diseases.
Collapse
Affiliation(s)
| | - Pradeep Reddy
- Altos Labs, San Diego Institute of Science, San Diego, CA, USA
| | | | | |
Collapse
|
7
|
St Pierre L, Berhan A, Sung EK, Alvarez JR, Wang H, Ji Y, Liu Y, Yu H, Meier A, Afshar K, Golts EM, Lin GY, Castaldi A, Calvert BA, Ryan A, Zhou B, Offringa IA, Marconett CN, Borok Z. Integrated multiomic analysis identifies TRIP13 as a mediator of alveolar epithelial type II cell dysfunction in idiopathic pulmonary fibrosis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167572. [PMID: 39547519 PMCID: PMC11951472 DOI: 10.1016/j.bbadis.2024.167572] [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: 04/14/2024] [Revised: 10/14/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a lethal progressive lung disease urgently needing new therapies. Current treatments only delay disease progression, leaving lung transplant as the sole remaining option. Recent studies support a model whereby IPF arises because alveolar epithelial type II (AT2) cells, which normally mediate distal lung regeneration, acquire airway and/or mesenchymal characteristics, preventing proper repair. Mechanisms driving this abnormal differentiation remain unclear. We performed integrated transcriptomic and epigenomic analysis of purified AT2 cells which revealed genome-wide alterations in IPF lungs. The most prominent epigenetic alteration was activation of an enhancer in thyroid receptor interactor 13 (TRIP13), although TRIP13 was not the most significantly transcriptionally upregulated gene. TRIP13 is broadly implicated in epithelial-mesenchymal plasticity. In cultured human AT2 cells and lung slices, small molecule TRIP13 inhibitor DCZ0415 prevented acquisition of the mesenchymal gene signature characteristic of IPF, suggesting TRIP13 inhibition as a potential therapeutic approach to fibrotic disease.
Collapse
Affiliation(s)
- Laurence St Pierre
- Department of Surgery, University of Southern California, Los Angeles, CA 90089, USA; Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Asres Berhan
- Department of Medicine, University of California San Diego, CA 92037, USA
| | - Eun K Sung
- Department of Translational Genomics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Department of Integrative Translational Sciences, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Juan R Alvarez
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Hongjun Wang
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Yanbin Ji
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Yixin Liu
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Haoze Yu
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Angela Meier
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA 92037, USA
| | - Kamyar Afshar
- Department of Medicine, University of California San Diego, CA 92037, USA
| | - Eugene M Golts
- Department of Surgery, University of California, San Diego, La Jolla, CA 92037, USA
| | - Grace Y Lin
- Department of Pathology, University of California, San Diego, La Jolla, CA 92037, USA
| | | | - Ben A Calvert
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Amy Ryan
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Beiyun Zhou
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Ite A Offringa
- Department of Surgery, University of Southern California, Los Angeles, CA 90089, USA; Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA.
| | - Crystal N Marconett
- Department of Surgery, University of Southern California, Los Angeles, CA 90089, USA; Department of Translational Genomics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Department of Integrative Translational Sciences, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA.
| | - Zea Borok
- Department of Medicine, University of California San Diego, CA 92037, USA.
| |
Collapse
|
8
|
Güler A, Yardımcı BK, Özek NŞ. Human anti-apoptotic Bcl-2 and Bcl-xL proteins protect yeast cells from aging induced oxidative stress. Biochimie 2025; 229:69-83. [PMID: 39413900 DOI: 10.1016/j.biochi.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 09/24/2024] [Accepted: 10/12/2024] [Indexed: 10/18/2024]
Abstract
Aging is a degenerative, biological, and time-dependent process that affects all organisms. Yeast aging is a physiological phenomenon characterized by the progressive transformation of yeast cells, resulting in modifications to their viability and vitality. Aging in yeast cells is comparable to that in higher organisms in some respects; however, due to their straightforward and well-characterized genetic makeup, these cells present unique advantages when it comes to researching the aging process. Here, we assessed the impact of human anti-apoptotic Bcl-2 and Bcl-xL proteins on aging using a yeast model. The findings clearly showed that these proteins exhibited remarkable anti-aging properties in yeast cells. Our data indicate that the presence of both proteins enhanced the reproductive survival of aging cells, likely by effecting the components functioning as both pro- and anti-oxidants, depending on the stage of yeast cell lifespan. Both proteins partially protected yeast cells from aging-related morphological deformations and cellular damage during the aging period. In particular, Bcl-xL expressing yeast cells reached the maximum activity levels for almost all of the major antioxidant enzymes and the total antioxidant status on the 8th day of lifespan and could provide effective protection at the latest stage of the investigated aging period. The chemometric data analysis of IR spectra confirmed the findings of the morphological and biochemical analyses. In this regard, specifically, understanding the mechanism of action on the cellular redox state of Bcl-xL in yeast may facilitate comprehension of its indirect antioxidant function in higher eukaryotes.
Collapse
Affiliation(s)
- Ayşenur Güler
- Chemistry Department, Graduate School of Natural and Applied Sciences, Pamukkale University, Denizli, Turkey
| | - Berna Kavakcıoğlu Yardımcı
- Department of Chemistry, Faculty of Science, Pamukkale University, Denizli, Turkey; Advanced Technology Application and Research Center, Pamukkale University, Denizli, Turkey.
| | - Nihal Şimşek Özek
- Department of Biology, Faculty of Science, Ataturk University, Erzurum, Turkey; East Anatolian High Technology Research and Application Center, Ataturk University, Erzurum, Turkey
| |
Collapse
|
9
|
Ruiz E, Leprieur F, Sposito G, Lüthi M, Schmidlin M, Panfili J, Pellissier L, Albouy C. Environmental DNA Epigenetics Accurately Predicts the Age of Cultured Fish Larvae. Ecol Evol 2025; 15:e70645. [PMID: 39944907 PMCID: PMC11821287 DOI: 10.1002/ece3.70645] [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: 10/23/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 02/19/2025] Open
Abstract
While acquiring age information is crucial for efficient stock management and biodiversity conservation, traditional aging methods fail to offer a universal, non-invasive, and precise way of estimating a wild animal's age. DNA methylation from tissue DNA (tDNA) was recently proposed as a method to overcome these issues and showed more accurate results than telomere-length-based age assessments. Here, we used environmental DNA (eDNA) for the first time as a template for age estimation, focusing on the larval phase (10-24 days post-hatch) of cultured Dicentrarchus labrax (seabass), a species of major economic and conservation interest. Using third-generation sequencing, we were able to directly detect various modification types (e.g., cytosine and adenosine methylation in all contexts) across the whole genome using amplification-free nanopore sequencing. However, aging sites were only present in the mitogenome, which could be a specific feature of eDNA methylation or the consequence of better DNA protection within mitochondria. By considering qualitative and quantitative information about aging sites according to an objective model selection framework, our epigenetic clock reached a cross-validated accuracy of 2.6 days (Median Absolute Error). Such performances are higher than those of previous clocks, notably for adult seabass even when scaling MAE to the age range, which could be linked to a more dynamic epigenome during early life stages. Overall, our pilot study proposes new methods to determine the potential of eDNA for simultaneous age and biodiversity assessments, although robust validation of our preliminary results along with methodological developments are needed before field applications can be envisaged.
Collapse
Affiliation(s)
- Eliot Ruiz
- MARBEC, Univ Montpellier, IRD, IFREMER, CNRSMontpellierFrance
| | - Fabien Leprieur
- MARBEC, Univ Montpellier, IRD, IFREMER, CNRSMontpellierFrance
| | - Gérard Sposito
- Mediterranean Coastal Environment StationUniversity of MontpellierSèteFrance
| | - Martina Lüthi
- Department of Environmental Systems Science, Ecosystems and Landscape Evolution, Institute of Terrestrial EcosystemsETH ZürichZürichSwitzerland
- Land Change Science Research UnitSwiss Federal Research Institute WSLBirmensdorfSwitzerland
| | - Michel Schmidlin
- Department of Environmental Systems Science, Ecosystems and Landscape Evolution, Institute of Terrestrial EcosystemsETH ZürichZürichSwitzerland
- Land Change Science Research UnitSwiss Federal Research Institute WSLBirmensdorfSwitzerland
| | - Jacques Panfili
- MARBEC, Univ Montpellier, IRD, IFREMER, CNRSMontpellierFrance
| | - Loïc Pellissier
- Department of Environmental Systems Science, Ecosystems and Landscape Evolution, Institute of Terrestrial EcosystemsETH ZürichZürichSwitzerland
- Land Change Science Research UnitSwiss Federal Research Institute WSLBirmensdorfSwitzerland
| | - Camille Albouy
- Department of Environmental Systems Science, Ecosystems and Landscape Evolution, Institute of Terrestrial EcosystemsETH ZürichZürichSwitzerland
- Land Change Science Research UnitSwiss Federal Research Institute WSLBirmensdorfSwitzerland
| |
Collapse
|
10
|
Rong Y, Wu Y, Chen Y, Liu Q, Ai L, Wu Y, Zhu Y, Zhang Y, Liu C, Ma Y, Tong X, Jin J, Li X, Zhou Y, Ji S, Zhang S, Fan H. ZAR1/2-Regulated Epigenetic Modifications are Essential for Age-Associated Oocyte Quality Maintenance and Zygotic Activation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410305. [PMID: 39755931 PMCID: PMC11848533 DOI: 10.1002/advs.202410305] [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: 08/26/2024] [Revised: 12/23/2024] [Indexed: 01/06/2025]
Abstract
The developmental competence and epigenetic progression of oocytes gradually become dysregulated with increasing maternal age. However, the mechanisms underlying age-related epigenetic regulation in oocytes remain poorly understood. Zygote arrest proteins 1 and 2 (ZAR1/2) are two maternal factors with partially redundant roles in maintaining oocyte quality, mainly known by regulating mRNA stability. In addition to this known function, it is found that ZAR1/2 is required for oocyte epigenetic maturation and zygotic reprogramming. Zar1/2-deleted oocytes exhibited reduced levels of multiple histone modifications and of the expression of corresponding histone modifiers, along with over-condensed chromatin, leading to compromised minor zygotic genome activation and deficient embryo development following fertilization. Cytoplasmic ZAR1/2 participated in intranuclear epigenetic maturation by binding the transcripts encoding histone modifiers and regulating their stability and translational activity. Moreover, oocytes from aged mice exhibited similar histone-modification deficiencies as the Zar1/2-deleted oocytes. ZAR1/2 mRNA and protein levels are downregulated in oocytes from mice and women with advanced ages, suggesting ZAR1/2 as regulators of epigenetic changes with reproductive aging. This study presents a new nucleo-cytoplasmic interaction mechanism that is involved in preventing oocyte epigenetic aging. Further, ZAR1/2 represents potential gene targets for diagnosis and clinical interventions in age-associated deficiencies in oocyte and embryo development.
Collapse
Affiliation(s)
- Yan Rong
- Department of Obstetrics and GynecologyZhejiang Key Laboratory of Precise Protection and Promotion of FertilityZhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseAssisted Reproduction UnitSir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310016China
- MOE Key Laboratory for Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences InstituteZhejiang UniversityHangzhou310058China
| | - Yu‐Ke Wu
- MOE Key Laboratory for Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences InstituteZhejiang UniversityHangzhou310058China
| | - Yingyan Chen
- Department of Obstetrics and GynecologyZhejiang Key Laboratory of Precise Protection and Promotion of FertilityZhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseAssisted Reproduction UnitSir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310016China
| | - Qing Liu
- Department of Traditional Chinese MedicineSir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Leilei Ai
- Department of Obstetrics and GynecologyZhejiang Key Laboratory of Precise Protection and Promotion of FertilityZhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseAssisted Reproduction UnitSir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310016China
| | - Yun‐Wen Wu
- MOE Key Laboratory for Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences InstituteZhejiang UniversityHangzhou310058China
| | - Yezhang Zhu
- MOE Key Laboratory for Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences InstituteZhejiang UniversityHangzhou310058China
| | - Yin‐Li Zhang
- Department of Obstetrics and GynecologyZhejiang Key Laboratory of Precise Protection and Promotion of FertilityZhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseAssisted Reproduction UnitSir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310016China
| | - Chengkan Liu
- Department of Obstetrics and GynecologyZhejiang Key Laboratory of Precise Protection and Promotion of FertilityZhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseAssisted Reproduction UnitSir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310016China
| | - Yerong Ma
- Department of Obstetrics and GynecologyZhejiang Key Laboratory of Precise Protection and Promotion of FertilityZhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseAssisted Reproduction UnitSir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310016China
| | - Xiaomei Tong
- Department of Obstetrics and GynecologyZhejiang Key Laboratory of Precise Protection and Promotion of FertilityZhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseAssisted Reproduction UnitSir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310016China
| | - Jiamin Jin
- Department of Obstetrics and GynecologyZhejiang Key Laboratory of Precise Protection and Promotion of FertilityZhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseAssisted Reproduction UnitSir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310016China
| | - Xiaoxuan Li
- Department of Obstetrics and GynecologyZhejiang Key Laboratory of Precise Protection and Promotion of FertilityZhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseAssisted Reproduction UnitSir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310016China
| | - Yan Zhou
- Department of Obstetrics and GynecologyZhejiang Key Laboratory of Precise Protection and Promotion of FertilityZhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseAssisted Reproduction UnitSir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310016China
| | - Shu‐Yan Ji
- MOE Key Laboratory for Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences InstituteZhejiang UniversityHangzhou310058China
| | - Songying Zhang
- Department of Obstetrics and GynecologyZhejiang Key Laboratory of Precise Protection and Promotion of FertilityZhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseAssisted Reproduction UnitSir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310016China
| | - Heng‐Yu Fan
- Department of Obstetrics and GynecologyZhejiang Key Laboratory of Precise Protection and Promotion of FertilityZhejiang Provincial Clinical Research Center for Reproductive Health and DiseaseAssisted Reproduction UnitSir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhou310016China
- MOE Key Laboratory for Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences InstituteZhejiang UniversityHangzhou310058China
- Center for Biomedical ResearchShaoxing InstituteZhejiang UniversityShaoxing312000China
| |
Collapse
|
11
|
Singh K, Jin Y, Hu MW, Palazzo I, Cano M, Hoang T, Bhutto I, Wang S, Sinha D, Blackshaw S, Qian J, Handa JT. Cigarette smoke and biological age induce degenerative heterogeneity in retinal pigment epithelium. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.27.635096. [PMID: 39974955 PMCID: PMC11838378 DOI: 10.1101/2025.01.27.635096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Environmental exposure such as cigarette smoke induces epigenetic changes that can induce degenerative heterogeneity and accelerate aging. In early age-related macular degeneration (AMD), the leading worldwide cause of blindness among the elderly, retinal pigment epithelial (RPE) cell heterogeneity is a key change. Since smoking is the strongest environmental risk factor for AMD, we hypothesized that cigarette smoke induces degenerative RPE heterogeneity through epigenetic changes that are distinct from aging, and that with aging, the RPE becomes vulnerable to cigarette smoke insult. We administered cigarette smoke condensate (CSC) intravitreally to young and aged mice and performed snRNA-seq and snATAC-seq on the RPE/choroid. This analysis identified separate cell clusters corresponding to healthy and abnormal, dedifferentiated RPE in both aged vehicle-treated and young CSC-treated mice. The dedifferentiated RPE were characterized by a global decrease in chromatin accessibility and decreased expression of genes in functional categories that were linked to hallmarks of aging. Notably, young, dedifferentiated RPE also exhibited a compensatory upregulation of hallmarks of aging-related genes, specifically those related to mitochondrial function and proteostasis. In contrast, aged dedifferentiated RPE did not express these compensatory changes, and did not survive CSC treatment, as experimentally verified with TUNEL labeling. These changes are relevant to early AMD because we identified through scRNA-seq, similar dedifferentiated and healthy macular RPE clusters in a donor who smoked and another with early AMD, but not from a nonsmoker. Degenerative cellular heterogeneity can include an abnormal cluster that jeopardizes cell survival and may represent an additional hallmark of ocular aging.
Collapse
Affiliation(s)
- Krishna Singh
- Department of Cell and Molecular Biology and Department of Ophthalmology, Tulane University, New Orleans, LA, 70118
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA 212872
| | - Yang Jin
- Department of Cell and Molecular Biology and Department of Ophthalmology, Tulane University, New Orleans, LA, 70118
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA 212872
| | - Ming-Wen Hu
- Department of Cell and Molecular Biology and Department of Ophthalmology, Tulane University, New Orleans, LA, 70118
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA 212872
| | - Isabella Palazzo
- Department of Cell and Molecular Biology and Department of Ophthalmology, Tulane University, New Orleans, LA, 70118
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA 212872
| | - Marisol Cano
- Department of Cell and Molecular Biology and Department of Ophthalmology, Tulane University, New Orleans, LA, 70118
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA 212872
| | - Thanh Hoang
- Department of Cell and Molecular Biology and Department of Ophthalmology, Tulane University, New Orleans, LA, 70118
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA 212872
| | - Imran Bhutto
- Department of Cell and Molecular Biology and Department of Ophthalmology, Tulane University, New Orleans, LA, 70118
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA 212872
| | - Shusheng Wang
- Department of Cell and Molecular Biology and Department of Ophthalmology, Tulane University, New Orleans, LA, 70118
| | - Debasish Sinha
- Department of Cell and Molecular Biology and Department of Ophthalmology, Tulane University, New Orleans, LA, 70118
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA 212872
| | - Seth Blackshaw
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA 212872
| | - Jiang Qian
- Department of Cell and Molecular Biology and Department of Ophthalmology, Tulane University, New Orleans, LA, 70118
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA 212872
| | - James T Handa
- Department of Cell and Molecular Biology and Department of Ophthalmology, Tulane University, New Orleans, LA, 70118
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21287
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA 212872
| |
Collapse
|
12
|
Zheng X, Xu Z, Zhao J, Hao S, Xu F, Ding S, Liu G, Yang S, Otsen B, Zhu W, Bai Z, Yang J, Chen R. Disparities in anxiety and related factors among Chinese older adults across different aged-care models: a comparison of two cross-sectional studies. BMC Geriatr 2025; 25:46. [PMID: 39838310 PMCID: PMC11748326 DOI: 10.1186/s12877-024-05653-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 12/23/2024] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Anxiety disorders in older adults have become a prominent public health problem due to their concomitant chronic conditions, reduced quality of life and even death. However, fewer studies have been conducted on differences in anxiety among older individuals in different aged-care models, and the interactive relationship between the influencing factors on anxiety remains unclear. The study aimed to examine the disparities in the prevalence of anxiety between community-dwelling and institutionalized older adults and related influencing factors. METHODS Data were collected from the Anhui Healthy Longevity Survey (AHLS) and the Anhui Elderly Caring Social Organizations Survey (AECSOS). Data on demographic variables, lifestyle factors and health-related variables in 6968 older adults were used for analysis. Anxiety symptoms were evaluated using the Generalized Anxiety Disorder Assessment Scale (GAD-7). Binary logistic regression models and a Classification and Regression Tree model (CART) were utilized to examine the relationship between variables. RESULTS The prevalence of anxiety were 24.3% and 16.7% among community-dwelling older adults and institutionalized older adults, respectively. Several factors including age, gender, residence, education, income level, live alone, and number of chronic diseases showed a linkage with anxiety among community-dwelling older adults. For the institutionalized older adults, gender, residence, source of income, and number of chronic diseases exhibited a significant association with anxiety. We noted the interactive effect, suggesting that community-dwelling female older adults with an income level of less than 6500 RMB per year and reported chronic disease comorbidities had the highest likelihood of anxiety, and institutionalized female older adults with income sources such as pension, subsidy, family providing, and resident in rural areas have the greatest risk of experiencing anxiety. CONCLUSIONS This study has brought to light the higher risk of anxiety among community-dwelling older adults compared to institutionalized older adults. Targeted interventions are, therefore emphasized to address the negative impact of anxiety for populations at higher risk.
Collapse
Affiliation(s)
- Xin Zheng
- School of Public Health, Anhui Medical University, Hefei, China
- Chaohu Hospital of Anhui Medical University, Hefei, 238000, China
- School of Health Services Management, Anhui Medical University, Hefei, China
| | - Ziwen Xu
- School of Health Services Management, Anhui Medical University, Hefei, China
| | - Jiajie Zhao
- School of Health Services Management, Anhui Medical University, Hefei, China
| | - Sanyuan Hao
- School of Health Services Management, Anhui Medical University, Hefei, China
| | - Fuqin Xu
- School of Health Services Management, Anhui Medical University, Hefei, China
| | - Shuo Ding
- School of Health Services Management, Anhui Medical University, Hefei, China
| | - Guoqing Liu
- School of Health Services Management, Anhui Medical University, Hefei, China
| | - Shufan Yang
- School of Mechanical Engineering, University of Leeds, Leeds, UK
| | - Benjamin Otsen
- School of Health Services Management, Anhui Medical University, Hefei, China
- Registrars' Department, University of Cape Coast, Cape Coast, Ghana
| | - Wen Zhu
- The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhongliang Bai
- School of Health Services Management, Anhui Medical University, Hefei, China
| | - Jie Yang
- Chaohu Hospital of Anhui Medical University, Hefei, 238000, China
| | - Ren Chen
- School of Public Health, Anhui Medical University, Hefei, China.
- Chaohu Hospital of Anhui Medical University, Hefei, 238000, China.
- School of Health Services Management, Anhui Medical University, Hefei, China.
| |
Collapse
|
13
|
Camacho P, Ribeiro E, Pereira B, Nascimento J, Caldeira Rosa P, Henriques J, Barrão S, Sadio S, Quendera B, Delgadinho M, Ginete C, Silva C, Brito M. DNA Methyltransferase Expression (DNMT1, DNMT3a, and DNMT3b) as a Potential Biomarker in Age-Related Macular Degeneration. J Clin Med 2025; 14:559. [PMID: 39860565 PMCID: PMC11765804 DOI: 10.3390/jcm14020559] [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: 12/10/2024] [Revised: 01/08/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Age-related macular degeneration (AMD) is a global cause of vision loss, with limited therapeutic options highlighting the need for effective biomarkers. This study aimed to characterize plasma DNA methyltransferase expression (DNMT1, DNMT3A, and DNMT3B) in AMD patients and explore divergent expression patterns across different stages of AMD. Methods: Thirty-eight AMD patients were prospectively enrolled and stratified by disease severity: eAMD, iAMD, nAMD, and aAMD. Comprehensive ophthalmological assessments were performed, including best-corrected visual acuity, digital color fundus photographs, and Spectral Domain Optical Coherence Tomography. Peripheral blood samples were collected for RNA extraction and qRT-PCR to access epigenetic effectors' transcriptional expression, namely DNMT1, DNMT3A, and DNMT3B genes. The collected data were analyzed using IBM SPSS 29. Results:DNMT1 expression was significantly downregulated in late AMD (-0.186 ± 0.341) compared to early/intermediate AMD (0.026 ± 0.246). Within late AMD, aAMD exhibited a marked downregulation of DNMT1 (-0.375 ± 0.047) compared to nAMD (0.129 ± 0.392). DNMT3A and DNMT3B showed similar divergent expression patterns, correlating with disease stage. Conclusions: This study identified stage-specific transcriptional differences in DNMT expression, emphasizing its potential as a biomarker for AMD progression and a target for future research into personalized therapeutic strategies.
Collapse
Affiliation(s)
- Pedro Camacho
- H&TRC—Health & Technology Research Center, ESTeSL—Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, 1990096 Lisbon, Portugal; (E.R.); (B.P.); (M.D.); (C.S.); (M.B.)
| | - Edna Ribeiro
- H&TRC—Health & Technology Research Center, ESTeSL—Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, 1990096 Lisbon, Portugal; (E.R.); (B.P.); (M.D.); (C.S.); (M.B.)
| | - Bruno Pereira
- H&TRC—Health & Technology Research Center, ESTeSL—Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, 1990096 Lisbon, Portugal; (E.R.); (B.P.); (M.D.); (C.S.); (M.B.)
- Retina Institute of Lisbon, 1150085 Lisbon, Portugal; (J.N.); (P.C.R.); (J.H.); (S.S.)
- iNOVA4Health, NOVA Medical School, NMS, Faculdade de Ciências Médicas, FCM, Universidade NOVA de Lisboa, 1169056 Lisboa, Portugal
| | - João Nascimento
- Retina Institute of Lisbon, 1150085 Lisbon, Portugal; (J.N.); (P.C.R.); (J.H.); (S.S.)
- Beatriz Ângelo Hospital, 2674514 Lisbon, Portugal
| | - Paulo Caldeira Rosa
- Retina Institute of Lisbon, 1150085 Lisbon, Portugal; (J.N.); (P.C.R.); (J.H.); (S.S.)
| | - José Henriques
- Retina Institute of Lisbon, 1150085 Lisbon, Portugal; (J.N.); (P.C.R.); (J.H.); (S.S.)
| | - Sandra Barrão
- Ophthalmology Department, Centro Hospitalar Universitário de Lisboa Central, 1150199 Lisbon, Portugal; (S.B.); (B.Q.)
| | - Silvia Sadio
- Retina Institute of Lisbon, 1150085 Lisbon, Portugal; (J.N.); (P.C.R.); (J.H.); (S.S.)
- Ophthalmology Department, Centro Hospitalar Universitário de Lisboa Central, 1150199 Lisbon, Portugal; (S.B.); (B.Q.)
| | - Bruno Quendera
- Ophthalmology Department, Centro Hospitalar Universitário de Lisboa Central, 1150199 Lisbon, Portugal; (S.B.); (B.Q.)
| | - Mariana Delgadinho
- H&TRC—Health & Technology Research Center, ESTeSL—Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, 1990096 Lisbon, Portugal; (E.R.); (B.P.); (M.D.); (C.S.); (M.B.)
| | - Catarina Ginete
- H&TRC—Health & Technology Research Center, ESTeSL—Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, 1990096 Lisbon, Portugal; (E.R.); (B.P.); (M.D.); (C.S.); (M.B.)
| | - Carina Silva
- H&TRC—Health & Technology Research Center, ESTeSL—Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, 1990096 Lisbon, Portugal; (E.R.); (B.P.); (M.D.); (C.S.); (M.B.)
| | - Miguel Brito
- H&TRC—Health & Technology Research Center, ESTeSL—Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, 1990096 Lisbon, Portugal; (E.R.); (B.P.); (M.D.); (C.S.); (M.B.)
| |
Collapse
|
14
|
Lv J, Chen Q, Wang J, Guo N, Fang Y, Guo Q, Li J, Ma X, Zhan H, Chen W, Wang L, Yan Q, Tong J, Wang Z. Downregulation of MLF1 safeguards cardiomyocytes against senescence-associated chromatin opening. Nucleic Acids Res 2025; 53:gkae1176. [PMID: 39657728 PMCID: PMC11754730 DOI: 10.1093/nar/gkae1176] [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: 11/05/2023] [Revised: 10/21/2024] [Accepted: 11/12/2024] [Indexed: 12/12/2024] Open
Abstract
Aging-associated cardiac hypertrophy (AACH) increases susceptibility to heart failure in the elderly. Chromatin remodeling contributes to the gene reprogramming in AACH; however, the intrinsic regulations remain elusive. We performed a transcriptome analysis for AACH in comparison with pressure-overload-induced pathological cardiac hypertrophy in mice and identified myeloid leukemia factor 1 (MLF1) as an aging-sensitive factor whose expression was reduced during aging but could be reversed by anti-aging administrations. In human AC16 cardiomyocytes, silencing MLF1 suppressed H2O2-induced cell senescence while the phenotype was exacerbated by MLF1 overexpression. RNA-seq analysis revealed that MLF1 functioned as a transcription activator, regulating genomic-clustered genes that mainly involved in inflammation and development. ATAC-seq analysis showed a prominent reduction in chromatin accessibility at the promoter regions of senescence effectors, like IL1B and p21, after MLF1 knockdown. Despite a potential interaction of MLF1 with the histone methyltransferase PRC2, its inhibition failed to reverse the impact of MLF1 knockdown. Instead, MLF1-mediated regulation was blunted by inhibiting the acetyltransferase EP300. CUT&Tag analysis showed that MLF1 bound to target promoters and recruited EP300 to promote H3K27ac deposition. Collectively, we identify MLF1 as a pro-aging epigenetic orchestrator that recruits EP300 to facilitate opening of the condensed chromatin encompassing senescence effectors.
Collapse
Affiliation(s)
- Jian Lv
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Shenzhen Hospital, Chinese Academy of Medical Sciences, Shenzhen 518057, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Qin Chen
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Shenzhen Hospital, Chinese Academy of Medical Sciences, Shenzhen 518057, China
- School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Junmei Wang
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Shenzhen Hospital, Chinese Academy of Medical Sciences, Shenzhen 518057, China
| | - Ningning Guo
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Shenzhen Hospital, Chinese Academy of Medical Sciences, Shenzhen 518057, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yu Fang
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Shenzhen Hospital, Chinese Academy of Medical Sciences, Shenzhen 518057, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Qiuxiao Guo
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Shenzhen Hospital, Chinese Academy of Medical Sciences, Shenzhen 518057, China
| | - Jiajie Li
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Shenzhen Hospital, Chinese Academy of Medical Sciences, Shenzhen 518057, China
| | - Xiao Ma
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Hongchao Zhan
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Shenzhen Hospital, Chinese Academy of Medical Sciences, Shenzhen 518057, China
| | - Weihao Chen
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Shenzhen Hospital, Chinese Academy of Medical Sciences, Shenzhen 518057, China
| | - Li Wang
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Shenzhen Hospital, Chinese Academy of Medical Sciences, Shenzhen 518057, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Qingqing Yan
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Shenzhen Hospital, Chinese Academy of Medical Sciences, Shenzhen 518057, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Jingjing Tong
- School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Zhihua Wang
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Shenzhen Hospital, Chinese Academy of Medical Sciences, Shenzhen 518057, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| |
Collapse
|
15
|
Mensah EO, Danyo EK, Asase RV. Exploring the effect of different diet types on ageing and age-related diseases. Nutrition 2025; 129:112596. [PMID: 39488864 DOI: 10.1016/j.nut.2024.112596] [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: 03/15/2024] [Revised: 08/21/2024] [Accepted: 09/30/2024] [Indexed: 11/05/2024]
Abstract
In recent times, there has been growing interest in understanding the factors contributing to prolonged and healthy lifespans observed in specific populations, tribes, or countries. Factors such as environmental and dietary play significant roles in shaping the ageing process and are often the focus of inquiries seeking to unravel the secrets behind longevity. Among these factors, diet emerges as a primary determinant, capable of either promoting or mitigating the onset of age-related diseases that impact the ageing trajectory. This review examines the impact of various diet types on ageing and age-related conditions, including cardiovascular disease, cancer, neurodegenerative disorders, and metabolic syndrome. Different dietary patterns, such as the Mediterranean diet, the Japanese diet, vegetarian and vegan diets, as well as low-carbohydrate and ketogenic diets, are evaluated for their potential effects on longevity and health span. Each diet type is characterized by distinct nutritional profiles, emphasizing specific food groups, macronutrient compositions, and bioactive components, which may exert diverse effects on ageing processes and disease risk. Additionally, dietary factors such as calorie restriction, intermittent fasting, and dietary supplementation are explored for their potential anti-ageing and disease-modifying effects. Understanding the influence of various diet types on ageing and age-related diseases can inform personalized dietary recommendations and lifestyle interventions aimed at promoting healthy aging and mitigating age-associated morbidities.
Collapse
Affiliation(s)
- Emmanuel O Mensah
- Faculty of Ecotechnology, ITMO University, Saint Petersburg, Russian Federation.
| | - Emmanuel K Danyo
- Institute of Chemical Engineering, Ural Federal University, Yekaterinburg, Russian Federation
| | - Richard V Asase
- Institute of Chemical Engineering, Ural Federal University, Yekaterinburg, Russian Federation
| |
Collapse
|
16
|
Wang Y, Zhang W, Zhang C, Van HQT, Seino T, Zhang Y. Reducing functionally defective old HSCs alleviates aging-related phenotypes in old recipient mice. Cell Res 2025; 35:45-58. [PMID: 39743633 PMCID: PMC11701126 DOI: 10.1038/s41422-024-01057-5] [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: 06/13/2024] [Accepted: 11/08/2024] [Indexed: 01/04/2025] Open
Abstract
Aging is a process accompanied by functional decline in tissues and organs with great social and medical consequences. Developing effective anti-aging strategies is of great significance. In this study, we demonstrated that transplantation of young hematopoietic stem cells (HSCs) into old mice can mitigate aging phenotypes, underscoring the crucial role of HSCs in the aging process. Through comprehensive molecular and functional analyses, we identified a subset of HSCs in aged mice that exhibit "younger" molecular profiles and functions, marked by low levels of CD150 expression. Mechanistically, CD150low HSCs from old mice but not their CD150high counterparts can effectively differentiate into downstream lineage cells. Notably, transplantation of old CD150low HSCs attenuates aging phenotypes and prolongs lifespan of elderly mice compared to those transplanted with unselected or CD150high HSCs. Importantly, reducing the dysfunctional CD150high HSCs can alleviate aging phenotypes in old recipient mice. Thus, our study demonstrates the presence of "younger" HSCs in old mice, and that aging-associated functional decline can be mitigated by reducing dysfunctional HSCs.
Collapse
Affiliation(s)
- Yuting Wang
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Wenhao Zhang
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Chao Zhang
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Hoang Q Tran Van
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Takashi Seino
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Yi Zhang
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA.
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA.
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA.
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Boston, MA, USA.
| |
Collapse
|
17
|
Zhuang X, Wang Q, Joost S, Ferrena A, Humphreys DT, Li Z, Blum M, Krause K, Ding S, Landais Y, Zhan Y, Zhao Y, Chaligne R, Lee JH, Carrasco SE, Bhanot UK, Koche RP, Bott MJ, Katajisto P, Soto-Feliciano YM, Pisanic T, Thomas T, Zheng D, Wong ES, Tammela T. Ageing limits stemness and tumorigenesis by reprogramming iron homeostasis. Nature 2025; 637:184-194. [PMID: 39633048 DOI: 10.1038/s41586-024-08285-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 10/24/2024] [Indexed: 12/07/2024]
Abstract
Ageing is associated with a decline in the number and fitness of adult stem cells1,2. Ageing-associated loss of stemness is posited to suppress tumorigenesis3,4, but this hypothesis has not been tested in vivo. Here we use physiologically aged autochthonous genetically engineered5,6 mouse models and primary cells5,6 to demonstrate that ageing suppresses lung cancer initiation and progression by degrading the stemness of the alveolar cell of origin. This phenotype is underpinned by the ageing-associated induction of the transcription factor NUPR1 and its downstream target lipocalin-2 in the cell of origin in mice and humans, which leads to functional iron insufficiency in the aged cells. Genetic inactivation of the NUPR1-lipocalin-2 axis or iron supplementation rescues stemness and promotes the tumorigenic potential of aged alveolar cells. Conversely, targeting the NUPR1-lipocalin-2 axis is detrimental to young alveolar cells through ferroptosis induction. Ageing-associated DNA hypomethylation at specific enhancer sites is associated with increased NUPR1 expression, which is recapitulated in young alveolar cells through DNA methylation inhibition. We uncover that ageing drives functional iron insufficiency that leads to loss of stemness and tumorigenesis but promotes resistance to ferroptosis. These findings have implications for the therapeutic modulation of cellular iron homeostasis in regenerative medicine and in cancer prevention. Furthermore, our findings are consistent with a model whereby most human cancers initiate at a young age, thereby highlighting the importance of directing cancer prevention efforts towards young individuals.
Collapse
Affiliation(s)
- Xueqian Zhuang
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Qing Wang
- Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
| | - Simon Joost
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alexander Ferrena
- Institute for Clinical and Translational Research, Albert Einstein College of Medicine, New York, NY, USA
| | - David T Humphreys
- Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
| | - Zhuxuan Li
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Graduate School of Medical Science, Weill Cornell Medicine, New York, NY, USA
| | - Melissa Blum
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Klavdija Krause
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Selena Ding
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yuna Landais
- Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
| | - Yingqian Zhan
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yang Zhao
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Ronan Chaligne
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joo-Hyeon Lee
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Sebastian E Carrasco
- Laboratory of Comparative Pathology, Weill Cornell Medicine, Memorial Sloan Kettering Cancer Center and Rockefeller University, New York, NY, USA
| | - Umeshkumar K Bhanot
- Pathology Core Facility, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Richard P Koche
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Matthew J Bott
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pekka Katajisto
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Yadira M Soto-Feliciano
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Thomas Pisanic
- Institute for NanoBioTechnology, Department of Oncology-Cancer Genetics and Epigenetics, Johns Hopkins University, Baltimore, MD, USA
| | - Tiffany Thomas
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Deyou Zheng
- Institute for Clinical and Translational Research, Albert Einstein College of Medicine, New York, NY, USA
- Departments of Genetics, Neurology, and Neuroscience, Albert Einstein College of Medicine, New York, NY, USA
| | - Emily S Wong
- Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
- School of Biotechnology and Biomolecular Sciences, UNSW, Sydney, New South Wales, Australia
| | - Tuomas Tammela
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
18
|
Pangrazzi L, Meryk A. Molecular and Cellular Mechanisms of Immunosenescence: Modulation Through Interventions and Lifestyle Changes. BIOLOGY 2024; 14:17. [PMID: 39857248 PMCID: PMC11760833 DOI: 10.3390/biology14010017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/17/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025]
Abstract
Immunosenescence, the age-related decline in immune function, is a complex biological process with profound implications for health and longevity. This phenomenon, characterized by alterations in both innate and adaptive immunity, increases susceptibility to infections, reduces vaccine efficacy, and contributes to the development of age-related diseases. At the cellular level, immunosenescence manifests as decreased production of naive T and B cells, accumulation of memory and senescent cells, thymic involution, and dysregulated cytokine production. Recent advances in molecular biology have shed light on the underlying mechanisms of immunosenescence, including telomere attrition, epigenetic alterations, mitochondrial dysfunction, and changes in key signaling pathways such as NF-κB and mTOR. These molecular changes lead to functional impairments in various immune cell types, altering their proliferative capacity, differentiation, and effector functions. Emerging research suggests that lifestyle factors may modulate the rate and extent of immunosenescence at both cellular and molecular levels. Physical activity, nutrition, stress management, and sleep patterns have been shown to influence immune cell function, inflammatory markers, and oxidative stress in older adults. This review provides a comprehensive analysis of the molecular and cellular mechanisms underlying immunosenescence and explores how lifestyle interventions may impact these processes. We will examine the current understanding of immunosenescence at the genomic, epigenomic, and proteomic levels, and discuss how various lifestyle factors can potentially mitigate or partially reverse aspects of immune aging. By integrating recent findings from immunology, gerontology, and molecular biology, we aim to elucidate the intricate interplay between lifestyle and immune aging at the molecular level, potentially informing future strategies for maintaining immune competence in aging populations.
Collapse
Affiliation(s)
- Luca Pangrazzi
- Institute for Biomedical Aging Research, Faculty of Biology, University of Innsbruck, 6020 Innsbruck, Austria;
| | - Andreas Meryk
- Department of Pediatrics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
19
|
Mrabti C, Yang N, Desdín-Micó G, Alonso-Calleja A, Vílchez-Acosta A, Pico S, Parras A, Piao Y, Schoenfeldt L, Luo S, Haghani A, Brooke RT, Maza MDC, Branchina C, Bignon Y, Maroun CY, von Meyenn F, Naveiras O, Horvath S, Sen P, Ocampo A. Loss of H3K9 trimethylation leads to premature aging. RESEARCH SQUARE 2024:rs.3.rs-4012025. [PMID: 39764087 PMCID: PMC11702797 DOI: 10.21203/rs.3.rs-4012025/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Aging is the major risk factor for most human diseases and represents a major socioeconomical challenge for modern societies. Despite its importance, the process of aging remains poorly understood. Epigenetic dysregulation has been proposed as a key driver of the aging process. Alterations in transcriptional networks and chromatin structure might be central to age-related functional decline. A prevalent feature described during aging is the overall reduction in heterochromatin, specifically marked by the loss of the repressive histone modification, histone 3 lysine 9 trimethylation (H3K9me3). However, the role of H3K9me3 in aging, especially in mammals, remains unclear. Here we show using a novel mouse strain, "TKOc", carrying a triple knockout of three methyltransferases responsible for H3K9me3 deposition, that the inducible loss of H3K9me3 in adulthood results in premature aging. TKOc mice exhibit reduced lifespan, lower body weight, increased frailty index, multi-organ degeneration, transcriptional changes with significant upregulation of transposable elements, and accelerated epigenetic age. Our data strongly supports the concept that the loss of epigenetic information might directly drives the aging process. These findings reveal the importance of epigenetic regulation in aging and suggest that interventions targeting epigenetic modifications could potentially slow down or reverse age-related decline. Understanding the molecular mechanisms underlying the process of aging will be crucial for developing novel therapeutic strategies that can delay the onset of age-associated diseases and preserve human health at old age specially in rapidly aging societies.
Collapse
Affiliation(s)
- Calida Mrabti
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Vaud, Switzerland
| | - Na Yang
- Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Gabriela Desdín-Micó
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Vaud, Switzerland
| | - Alejandro Alonso-Calleja
- Laboratory of Regenerative Hematopoiesis, Department of Biomedical Sciences, University of Lausanne, Switzerland
- Laboratory of Metabolic Signaling, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Alba Vílchez-Acosta
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Vaud, Switzerland
| | - Sara Pico
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Vaud, Switzerland
| | | | - Yulan Piao
- Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Lucas Schoenfeldt
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Vaud, Switzerland
- EPITERNA SA, Epalinges, Switzerland
| | - Siyuan Luo
- Departement of Health Sciences and Technology, ETH Zurich, Zurich
| | | | - Robert T. Brooke
- Epigenetic Clock Development, Foundation, Torrance, California, USA
| | - María del Carmen Maza
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Vaud, Switzerland
| | - Clémence Branchina
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Vaud, Switzerland
| | - Yohan Bignon
- INSERM U1242 Oncogenesis Stress Signaling, Centre de Lutte Contre le Cancer Eugène Marquis, University of Rennes, Rennes, France
| | - Céline Yacoub Maroun
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Vaud, Switzerland
| | | | - Olaia Naveiras
- Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, MD 21224, USA
- Laboratory of Regenerative Hematopoiesis, Department of Biomedical Sciences, University of Lausanne, Switzerland
| | - Steve Horvath
- Altos Labs, San Diego, CA, USA
- Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Payel Sen
- Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Alejandro Ocampo
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Vaud, Switzerland
- EPITERNA SA, Epalinges, Switzerland
| |
Collapse
|
20
|
Kwon SY, Park YJ. Function of NAD metabolism in white adipose tissue: lessons from mouse models. Adipocyte 2024; 13:2313297. [PMID: 38316756 PMCID: PMC10877972 DOI: 10.1080/21623945.2024.2313297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 01/22/2024] [Indexed: 02/07/2024] Open
Abstract
Nicotinamide Adenine Dinucleotide (NAD) is an endogenous substance in redox reactions and regulates various functions in metabolism. NAD and its precursors are known for their anti-ageing and anti-obesity properties and are mainly active in the liver and muscle. Boosting NAD+ through supplementation with the precursors, such as nicotinamide mononucleotide (NMN) or nicotinamide riboside (NR), enhances insulin sensitivity and circadian rhythm in the liver, and improves mitochondrial function in the muscle. Recent evidence has revealed that the adipose tissue could be another direct target of NAD supplementation by attenuating inflammation and fat accumulation. Moreover, murine studies with genetically modified models demonstrated that nicotinamide phosphoribosyltransferase (NAMPT), a NAD regulatory enzyme that synthesizes NMN, played a critical role in lipogenesis and lipolysis in an adipocyte-specific manner. The tissue-specific effects of NAD+ metabolic pathways indicate a potential of the NAD precursors to control metabolic stress particularly via focusing on adipose tissue. Therefore, this narrative review raises an importance of NAD metabolism in white adipose tissue (WAT) through a variety of studies using different mouse models.
Collapse
Affiliation(s)
- So Young Kwon
- Graduate Program in System Health and Engineering, Ewha Womans University, Seoul, Republic of Korea
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Republic of Korea
| | - Yoon Jung Park
- Graduate Program in System Health and Engineering, Ewha Womans University, Seoul, Republic of Korea
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
21
|
Swer PB, Kharbuli B, Syiem D, Sharma R. Age-related decline in the expression of BRG1, ATM and ATR are partially reversed by dietary restriction in the livers of female mice. Biogerontology 2024; 25:1025-1037. [PMID: 38970714 DOI: 10.1007/s10522-024-10117-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 06/26/2024] [Indexed: 07/08/2024]
Abstract
BRG1 (Brahma-related gene 1) is a member of the SWI/SNF (switch/sucrose nonfermentable) chromatin remodeling complex which utilizes the energy from ATP hydrolysis for its activity. In addition to its role of regulating the expression of a vast array of genes, BRG1 mediates DNA repair upon genotoxic stress and regulates senescence. During organismal ageing, there is accumulation of unrepaired/unrepairable DNA damage due to progressive breakdown of the DNA repair machinery. The present study investigates the expression level of BRG1 as a function of age in the liver of 5- and 21-month-old female mice. It also explores the impact of dietary restriction on BRG1 expression in the old (21-month) mice. Salient findings of the study are: Real-time PCR and Western blot analyses reveal that BRG1 levels are higher in 5-month-old mice but decrease significantly with age. Dietary restriction increases BRG1 expression in the 21-month-old mice, nearly restoring it to the level observed in the younger group. Similar expression patterns are observed for DNA damage response genes ATM (Ataxia Telangiectasia Mutated) and ATR (Ataxia Telangiectasia and Rad3-related) with the advancement in age and which appears to be modulated by dietary restriction. BRG1 transcriptionally regulates ATM as a function of age and dietary restriction. These results suggest that BRG1, ATM and ATR are downregulated as mice age, and dietary restriction can restore their expression. This implies that dietary restriction may play a crucial role in regulating BRG1 and related gene expression, potentially maintaining liver repair and metabolic processes as mice age.
Collapse
Affiliation(s)
- Pynskhem Bok Swer
- Department of Biochemistry, North-Eastern Hill University, Shillong, 793022, India
| | | | - Donkupar Syiem
- Department of Biochemistry, North-Eastern Hill University, Shillong, 793022, India
| | - Ramesh Sharma
- Department of Biochemistry, North-Eastern Hill University, Shillong, 793022, India.
| |
Collapse
|
22
|
Postberg J, Schubert MT, Nin V, Wagner L, Piefke M. A perspective on epigenomic aging processes in the human brain and their plasticity in patients with mental disorders - a systematic review. Neurogenetics 2024; 25:351-366. [PMID: 38967831 PMCID: PMC11534990 DOI: 10.1007/s10048-024-00771-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 06/28/2024] [Indexed: 07/06/2024]
Abstract
The debate surrounding nature versus nurture remains a central question in neuroscience, psychology, and in psychiatry, holding implications for both aging processes and the etiology of mental illness. Epigenetics can serve as a bridge between genetic predisposition and environmental influences, thus offering a potential avenue for addressing these questions. Epigenetic clocks, in particular, offer a theoretical framework for measuring biological age based on DNA methylation signatures, enabling the identification of disparities between biological and chronological age. This structured review seeks to consolidate current knowledge regarding the relationship between mental disorders and epigenetic age within the brain. Through a comprehensive literature search encompassing databases such as EBSCO, PubMed, and ClinicalTrials.gov, relevant studies were identified and analyzed. Studies that met inclusion criteria were scrutinized, focusing on those with large sample sizes, analyses of both brain tissue and blood samples, investigation of frontal cortex markers, and a specific emphasis on schizophrenia and depressive disorders. Our review revealed a paucity of significant findings, yet notable insights emerged from studies meeting specific criteria. Studies characterized by extensive sample sizes, analysis of brain tissue and blood samples, assessment of frontal cortex markers, and a focus on schizophrenia and depressive disorders yielded particularly noteworthy results. Despite the limited number of significant findings, these studies shed light on the complex interplay between epigenetic aging and mental illness. While the current body of literature on epigenetic aging in mental disorders presents limited significant findings, it underscores the importance of further research in this area. Future studies should prioritize large sample sizes, comprehensive analyses of brain tissue and blood samples, exploration of specific brain regions such as the frontal cortex, and a focus on key mental disorders. Such endeavors will contribute to a deeper understanding of the relationship between epigenetic aging and mental illness, potentially informing novel diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Jan Postberg
- Clinical Molecular Genetics and Epigenetics, Faculty of Health, Witten/Herdecke University, Alfred-Herrhausen-Str. 50, 58448, Witten, Germany.
- Centre for Biomedical Education & Research (ZBAF), Witten/Herdecke University, Alfred-Herrhausen-Str. 50, 58448, Witten, Germany.
| | - Michèle Tina Schubert
- Neurobiology and Genetics of Behavior, Department of Psychology and Psychotherapy, Faculty of Health, Witten/Herdecke University, Alfred-Herrhausen-Str. 50, 58448, Witten, Germany
| | - Vincent Nin
- Neurobiology and Genetics of Behavior, Department of Psychology and Psychotherapy, Faculty of Health, Witten/Herdecke University, Alfred-Herrhausen-Str. 50, 58448, Witten, Germany
| | - Lukas Wagner
- Neurobiology and Genetics of Behavior, Department of Psychology and Psychotherapy, Faculty of Health, Witten/Herdecke University, Alfred-Herrhausen-Str. 50, 58448, Witten, Germany
| | - Martina Piefke
- Centre for Biomedical Education & Research (ZBAF), Witten/Herdecke University, Alfred-Herrhausen-Str. 50, 58448, Witten, Germany
- Neurobiology and Genetics of Behavior, Department of Psychology and Psychotherapy, Faculty of Health, Witten/Herdecke University, Alfred-Herrhausen-Str. 50, 58448, Witten, Germany
| |
Collapse
|
23
|
Fisher RMA, Torrente MP. Histone post-translational modification and heterochromatin alterations in neurodegeneration: revealing novel disease pathways and potential therapeutics. Front Mol Neurosci 2024; 17:1456052. [PMID: 39346681 PMCID: PMC11427407 DOI: 10.3389/fnmol.2024.1456052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 08/20/2024] [Indexed: 10/01/2024] Open
Abstract
Alzheimer's disease (AD), Parkinson's disease (PD), Frontotemporal Dementia (FTD), and Amyotrophic lateral sclerosis (ALS) are complex and fatal neurodegenerative diseases. While current treatments for these diseases do alleviate some symptoms, there is an imperative need for novel treatments able to stop their progression. For all of these ailments, most cases occur sporadically and have no known genetic cause. Only a small percentage of patients bear known mutations which occur in a multitude of genes. Hence, it is clear that genetic factors alone do not explain disease occurrence. Chromatin, a DNA-histone complex whose basic unit is the nucleosome, is divided into euchromatin, an open form accessible to the transcriptional machinery, and heterochromatin, which is closed and transcriptionally inactive. Protruding out of the nucleosome, histone tails undergo post-translational modifications (PTMs) including methylation, acetylation, and phosphorylation which occur at specific residues and are connected to different chromatin structural states and regulate access to transcriptional machinery. Epigenetic mechanisms, including histone PTMs and changes in chromatin structure, could help explain neurodegenerative disease processes and illuminate novel treatment targets. Recent research has revealed that changes in histone PTMs and heterochromatin loss or gain are connected to neurodegeneration. Here, we review evidence for epigenetic changes occurring in AD, PD, and FTD/ALS. We focus specifically on alterations in the histone PTMs landscape, changes in the expression of histone modifying enzymes and chromatin remodelers as well as the consequences of these changes in heterochromatin structure. We also highlight the potential for epigenetic therapies in neurodegenerative disease treatment. Given their reversibility and pharmacological accessibility, epigenetic mechanisms provide a promising avenue for novel treatments. Altogether, these findings underscore the need for thorough characterization of epigenetic mechanisms and chromatin structure in neurodegeneration.
Collapse
Affiliation(s)
- Raven M. A. Fisher
- PhD. Program in Biochemistry, City University of New York - The Graduate Center, New York, NY, United States
| | - Mariana P. Torrente
- Department of Chemistry and Biochemistry, Brooklyn College, Brooklyn, NY, United States
- PhD. Programs in Chemistry, Biochemistry, and Biology, City University of New York - The Graduate Center, New York, NY, United States
| |
Collapse
|
24
|
Liang Z, Chen S, Li Y, Lai W, Wang H. Adenosine Deaminase-Like Gene-Carried Lentivirus Toolkit for Identification of DNA N 6-Methyladenine Origins. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403376. [PMID: 39023073 PMCID: PMC11425204 DOI: 10.1002/advs.202403376] [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: 04/10/2024] [Revised: 06/25/2024] [Indexed: 07/20/2024]
Abstract
Post-replicative DNA N6-methyladenine (pr6mdA) can form via bona fide methylase-catalyzed adenine methylation, playing a pivotal role in embryonic development and other biological processes. Surprisingly, pre-methylated adenine can be erroneously incorporated into DNA as misincorporated N6-methyladenine (i6mdA) via DNA polymerase-mediated replication. Despite pr6mdA and i6mdA sharing identical chemical structures, their biological functions diverge significantly, presenting a substantial challenge in distinguishing between the two. Here, for the first-time, it is exploited that the adenosine deaminase-like (Adal) protein and a corresponding activity-null mutant to construct an Adal lentivirus toolkit. With this newly designed toolkit, both pr6mdA and i6mdA can be identified and quantified simultaneously. The presence of 6mdA in the bone marrow cells of mice is shown, with its levels serving as indicators for growth with age, probably reflecting the cellular stress-caused changes in RNA decay, nucleotide pool sanitation, and transcription. Collectively, a powerful toolkit to advance understanding of both pr6mdA and i6mdA is demonstrated.
Collapse
Affiliation(s)
- Ziyu Liang
- The State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 10085, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Shaokun Chen
- The State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 10085, P. R. China
| | - Yao Li
- The State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 10085, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Weiyi Lai
- The State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 10085, P. R. China
| | - Hailin Wang
- The State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 10085, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- School of Environment and Health, Jianghan University, Wuhan, 430056, China
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| |
Collapse
|
25
|
Izadi M, Sadri N, Abdi A, Serajian S, Jalalei D, Tahmasebi S. Epigenetic biomarkers in aging and longevity: Current and future application. Life Sci 2024; 351:122842. [PMID: 38879158 DOI: 10.1016/j.lfs.2024.122842] [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: 12/29/2023] [Revised: 06/06/2024] [Accepted: 06/12/2024] [Indexed: 06/20/2024]
Abstract
The aging process has been one of the most necessary research fields in the current century, and knowing different theories of aging and the role of different genetic, epigenetic, molecular, and environmental modulating factors in increasing the knowledge of aging mechanisms and developing appropriate diagnostic, therapeutic, and preventive ways would be helpful. One of the most conserved signs of aging is epigenetic changes, including DNA methylation, histone modifications, chromatin remodeling, noncoding RNAs, and extracellular RNAs. Numerous biological processes and hallmarks are vital in aging development, but epigenomic alterations are especially notable because of their importance in gene regulation and cellular identity. The mounting evidence points to a possible interaction between age-related epigenomic alterations and other aging hallmarks, like genome instability. To extend a healthy lifespan and possibly reverse some facets of aging and aging-related diseases, it will be crucial to comprehend global and locus-specific epigenomic modifications and recognize corresponding regulators of health and longevity. In the current study, we will aim to discuss the role of epigenomic mechanisms in aging and the most recent developments in epigenetic diagnostic biomarkers, which have the potential to focus efforts on reversing the destructive signs of aging and extending the lifespan.
Collapse
Affiliation(s)
- Mehran Izadi
- Department of Infectious and Tropical Diseases, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Synapse Laboratory Diagnostic Technologies Accelerator, Tehran, Iran; Department of Research & Technology, Zeenome Longevity Research Institute, Tehran, Iran
| | - Nariman Sadri
- Synapse Laboratory Diagnostic Technologies Accelerator, Tehran, Iran; Department of Research & Technology, Zeenome Longevity Research Institute, Tehran, Iran; School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirhossein Abdi
- Synapse Laboratory Diagnostic Technologies Accelerator, Tehran, Iran; Department of Research & Technology, Zeenome Longevity Research Institute, Tehran, Iran; Royan Institute for Stem Cell Biology and Technology, Tehran, Iran
| | - Sahar Serajian
- Synapse Laboratory Diagnostic Technologies Accelerator, Tehran, Iran; Department of Research & Technology, Zeenome Longevity Research Institute, Tehran, Iran; Royan Institute for Stem Cell Biology and Technology, Tehran, Iran
| | - Dorsa Jalalei
- Synapse Laboratory Diagnostic Technologies Accelerator, Tehran, Iran; Department of Research & Technology, Zeenome Longevity Research Institute, Tehran, Iran; School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Safa Tahmasebi
- Synapse Laboratory Diagnostic Technologies Accelerator, Tehran, Iran; Department of Research & Technology, Zeenome Longevity Research Institute, Tehran, Iran; Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
26
|
Zhang L, Kong D, Zhao X, Meng Y, Li J, Wang Z, Chai W. Tetrabromobisphenol S (TBBPS) exposure causes gastric cell senescence and inflammation by inducing iron overload. Toxicology 2024; 506:153866. [PMID: 38909936 DOI: 10.1016/j.tox.2024.153866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 06/06/2024] [Accepted: 06/14/2024] [Indexed: 06/25/2024]
Abstract
Tetrabromobisphenol S (TBBPS) is a brominated flame retardants (BFRs). TBBPS is widely used as a new type of BFR to replace TBBPA. Here, we used gastric cells as a model for evaluating the effect of TBBPS on the toxicology of gastric cells. Biochemical assays such as indirect immunofluorescence, cell proliferation assay were performed to analyze the toxicological effects of TBBPS on gastric cells. Cell proliferation analysis showed that TBBPS caused inhibition of gastric cell proliferation, and TBBPS induced gastric cell death. Further analysis showed that TBBPS led to ferroptosis and senescence of gastric cells by detecting ferroptosis-related marker molecules. Further work showed that TBBPS treatment resulted in lowered ferritin expression alongside heightened transferrin levels, which may be a potential molecular mechanism for TBBPS-induced ferroptosis and senescence in gastric cells. Here, our team investigates the effects of TBBPS on gastric cells in an in vitro model, and found that TBBPS caused toxicological damage to gastric cells. This study indicates potential toxic effects of TBBPS on the gastric cells, thereby providing a basis for further research into the toxicology of TBBPS.
Collapse
Affiliation(s)
- Lei Zhang
- The first department of Hepatobiliary and Pancreatic Surgery, Cangzhou Central Hospital, Cangzhou, China
| | - Deshuai Kong
- The first department of Hepatobiliary and Pancreatic Surgery, Cangzhou Central Hospital, Cangzhou, China
| | - Xiulei Zhao
- The first department of Hepatobiliary and Pancreatic Surgery, Cangzhou Central Hospital, Cangzhou, China
| | - Yu Meng
- The first department of Hepatobiliary and Pancreatic Surgery, Cangzhou Central Hospital, Cangzhou, China
| | - Jinchao Li
- The first department of Hepatobiliary and Pancreatic Surgery, Cangzhou Central Hospital, Cangzhou, China
| | - Zhenyong Wang
- The first department of Hepatobiliary and Pancreatic Surgery, Cangzhou Central Hospital, Cangzhou, China
| | - Wei Chai
- The first department of Hepatobiliary and Pancreatic Surgery, Cangzhou Central Hospital, Cangzhou, China.
| |
Collapse
|
27
|
Occean JR, Yang N, Sun Y, Dawkins MS, Munk R, Belair C, Dar S, Anerillas C, Wang L, Shi C, Dunn C, Bernier M, Price NL, Kim JS, Cui CY, Fan J, Bhattacharyya M, De S, Maragkakis M, de Cabo R, Sidoli S, Sen P. Gene body DNA hydroxymethylation restricts the magnitude of transcriptional changes during aging. Nat Commun 2024; 15:6357. [PMID: 39069555 PMCID: PMC11284234 DOI: 10.1038/s41467-024-50725-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 07/15/2024] [Indexed: 07/30/2024] Open
Abstract
DNA hydroxymethylation (5hmC), the most abundant oxidative derivative of DNA methylation, is typically enriched at enhancers and gene bodies of transcriptionally active and tissue-specific genes. Although aberrant genomic 5hmC has been implicated in age-related diseases, its functional role in aging remains unknown. Here, using mouse liver and cerebellum as model organs, we show that 5hmC accumulates in gene bodies associated with tissue-specific function and restricts the magnitude of gene expression changes with age. Mechanistically, 5hmC decreases the binding of splicing associated factors and correlates with age-related alternative splicing events. We found that various age-related contexts, such as prolonged quiescence and senescence, drive the accumulation of 5hmC with age. We provide evidence that this age-related transcriptionally restrictive function is conserved in mouse and human tissues. Our findings reveal that 5hmC regulates tissue-specific function and may play a role in longevity.
Collapse
Affiliation(s)
- James R Occean
- Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Na Yang
- Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Yan Sun
- Department of Biochemistry, Albert Einstein School of Medicine, Bronx, NY, USA
| | - Marshall S Dawkins
- Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Rachel Munk
- Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Cedric Belair
- Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Showkat Dar
- Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Carlos Anerillas
- Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Lin Wang
- Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Changyou Shi
- Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Christopher Dunn
- Flow Cytometry Unit, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Michel Bernier
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Nathan L Price
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Julie S Kim
- Department of Biochemistry, Albert Einstein School of Medicine, Bronx, NY, USA
| | - Chang-Yi Cui
- Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Jinshui Fan
- Computational Biology and Genomics Core, Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, MD, USA
| | | | - Supriyo De
- Computational Biology and Genomics Core, Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Manolis Maragkakis
- Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein School of Medicine, Bronx, NY, USA
| | - Payel Sen
- Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, MD, USA.
| |
Collapse
|
28
|
Shim HS, Iaconelli J, Shang X, Li J, Lan ZD, Jiang S, Nutsch K, Beyer BA, Lairson LL, Boutin AT, Bollong MJ, Schultz PG, DePinho RA. TERT activation targets DNA methylation and multiple aging hallmarks. Cell 2024; 187:4030-4042.e13. [PMID: 38908367 PMCID: PMC11552617 DOI: 10.1016/j.cell.2024.05.048] [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: 04/17/2023] [Revised: 04/03/2024] [Accepted: 05/23/2024] [Indexed: 06/24/2024]
Abstract
Insufficient telomerase activity, stemming from low telomerase reverse transcriptase (TERT) gene transcription, contributes to telomere dysfunction and aging pathologies. Besides its traditional function in telomere synthesis, TERT acts as a transcriptional co-regulator of genes pivotal in aging and age-associated diseases. Here, we report the identification of a TERT activator compound (TAC) that upregulates TERT transcription via the MEK/ERK/AP-1 cascade. In primary human cells and naturally aged mice, TAC-induced elevation of TERT levels promotes telomere synthesis, blunts tissue aging hallmarks with reduced cellular senescence and inflammatory cytokines, and silences p16INK4a expression via upregulation of DNMT3B-mediated promoter hypermethylation. In the brain, TAC alleviates neuroinflammation, increases neurotrophic factors, stimulates adult neurogenesis, and preserves cognitive function without evident toxicity, including cancer risk. Together, these findings underscore TERT's critical role in aging processes and provide preclinical proof of concept for physiological TERT activation as a strategy to mitigate multiple aging hallmarks and associated pathologies.
Collapse
Affiliation(s)
- Hong Seok Shim
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jonathan Iaconelli
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Xiaoying Shang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jiexi Li
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zheng D Lan
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shan Jiang
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kayla Nutsch
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Brittney A Beyer
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Luke L Lairson
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Adam T Boutin
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael J Bollong
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Peter G Schultz
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ronald A DePinho
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
29
|
Mrabti C, Yang N, Desdín-Micó G, Alonso-Calleja A, Vílchez-Acosta A, Pico S, Parras A, Piao Y, Schoenfeldt L, Luo S, Haghani A, Brooke R, del Carmen Maza M, Branchina C, Maroun CY, von Meyenn F, Naveiras O, Horvath S, Sen P, Ocampo A. Loss of H3K9 trimethylation leads to premature aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.24.604929. [PMID: 39091811 PMCID: PMC11291141 DOI: 10.1101/2024.07.24.604929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Aging is the major risk factor for most human diseases and represents a major socio-economical challenge for modern societies. Despite its importance, the process of aging remains poorly understood. Epigenetic dysregulation has been proposed as a key driver of the aging process. Modifications in transcriptional networks and chromatin structure might be central to age-related functional decline. A prevalent feature described during aging is the overall reduction in heterochromatin, specifically marked by the loss of repressive histone modification, Histone 3 lysine 9 trimethylation (H3K9me3). However, the role of H3K9me3 in aging, especially in mammals, remains unclear. Here we show using a novel mouse strain, (TKOc), carrying a triple knockout of three methyltransferases responsible for H3K9me3 deposition, that the inducible loss of H3K9me3 in adulthood results in premature aging. TKOc mice exhibit reduced lifespan, lower body weight, increased frailty index, multi-organ degeneration, transcriptional changes with significant upregulation of transposable elements, and accelerated epigenetic age. Our data strongly supports the concept that the loss of epigenetic information directly drives the aging process. These findings reveal the importance of epigenetic regulation in aging and suggest that interventions targeting epigenetic modifications could potentially slow down or reverse age-related decline. Understanding the molecular mechanisms underlying the process of aging will be crucial for developing novel therapeutic strategies that can delay the onset of age-associated diseases and preserve human health at old age specially in rapidly aging societies.
Collapse
Affiliation(s)
- Calida Mrabti
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Vaud, Switzerland
| | - Na Yang
- Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Gabriela Desdín-Micó
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Vaud, Switzerland
| | - Alejandro Alonso-Calleja
- Laboratory of Regenerative Hematopoiesis, Department of Biomedical Sciences, University of Lausanne, Switzerland
- Laboratory of Metabolic Signaling, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Alba Vílchez-Acosta
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Vaud, Switzerland
| | - Sara Pico
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Vaud, Switzerland
| | | | - Yulan Piao
- Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Lucas Schoenfeldt
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Vaud, Switzerland
- EPITERNA SA, Epalinges, Switzerland
| | - Siyuan Luo
- Departement of Health Sciences and Technology, ETH Zurich, Zurich
| | | | - Robert Brooke
- Epigenetic Clock Development, Foundation, Torrance, California, USA
| | - María del Carmen Maza
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Vaud, Switzerland
| | - Clémence Branchina
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Vaud, Switzerland
| | - Céline Yacoub Maroun
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Vaud, Switzerland
| | | | - Olaia Naveiras
- Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, MD 21224, USA
- Laboratory of Regenerative Hematopoiesis, Department of Biomedical Sciences, University of Lausanne, Switzerland
| | - Steve Horvath
- Altos Labs, San Diego, CA, USA
- Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Payel Sen
- Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Alejandro Ocampo
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Vaud, Switzerland
- EPITERNA SA, Epalinges, Switzerland
| |
Collapse
|
30
|
Occean JR, Yang N, Sun Y, Dawkins MS, Munk R, Belair C, Dar S, Anerillas C, Wang L, Shi C, Dunn C, Bernier M, Price NL, Kim JS, Cui CY, Fan J, Bhattacharyya M, De S, Maragkakis M, deCabo R, Sidoli S, Sen P. Gene body DNA hydroxymethylation restricts the magnitude of transcriptional changes during aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.02.15.528714. [PMID: 36824863 PMCID: PMC9949049 DOI: 10.1101/2023.02.15.528714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
DNA hydroxymethylation (5hmC), the most abundant oxidative derivative of DNA methylation, is typically enriched at enhancers and gene bodies of transcriptionally active and tissue-specific genes. Although aberrant genomic 5hmC has been implicated in age-related diseases, its functional role in aging remains unknown. Here, using mouse liver and cerebellum as model organs, we show that 5hmC accumulates in gene bodies associated with tissue-specific function and restricts the magnitude of gene expression changes with age. Mechanistically, 5hmC decreases the binding of splicing associated factors and correlates with age-related alternative splicing events. We found that various age-related contexts, such as prolonged quiescence and senescence, drive the accumulation of 5hmC with age. We provide evidence that this age-related transcriptionally restrictive function is conserved in mouse and human tissues. Our findings reveal that 5hmC regulates tissue-specific function and may play a role in longevity.
Collapse
|
31
|
Ma R, Zhang Y, Zhang J, Zhang P, Liu Z, Fan Y, Wang HT, Zhang Z, Zhu B. Targeting pericentric non-consecutive motifs for heterochromatin initiation. Nature 2024; 631:678-685. [PMID: 38961301 DOI: 10.1038/s41586-024-07640-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 05/31/2024] [Indexed: 07/05/2024]
Abstract
Pericentric heterochromatin is a critical component of chromosomes marked by histone H3 K9 (H3K9) methylation1-3. However, what recruits H3K9-specific histone methyltransferases to pericentric regions in vertebrates remains unclear4, as does why pericentric regions in different species share the same H3K9 methylation mark despite lacking highly conserved DNA sequences2,5. Here we show that zinc-finger proteins ZNF512 and ZNF512B specifically localize at pericentric regions through direct DNA binding. Notably, both ZNF512 and ZNF512B are sufficient to initiate de novo heterochromatin formation at ectopically targeted repetitive regions and pericentric regions, as they directly recruit SUV39H1 and SUV39H2 (SUV39H) to catalyse H3K9 methylation. SUV39H2 makes a greater contribution to H3K9 trimethylation, whereas SUV39H1 seems to contribute more to silencing, probably owing to its preferential association with HP1 proteins. ZNF512 and ZNF512B from different species can specifically target pericentric regions of other vertebrates, because the atypical long linker residues between the zinc-fingers of ZNF512 and ZNF512B offer flexibility in recognition of non-consecutively organized three-nucleotide triplets targeted by each zinc-finger. This study addresses two long-standing questions: how constitutive heterochromatin is initiated and how seemingly variable pericentric sequences are targeted by the same set of conserved machinery in vertebrates.
Collapse
Affiliation(s)
- Runze Ma
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Epigenetic Regulation and Intervention, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yan Zhang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Epigenetic Regulation and Intervention, Chinese Academy of Sciences, Beijing, China
| | - Jing Zhang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Epigenetic Regulation and Intervention, Chinese Academy of Sciences, Beijing, China
| | - Pinqi Zhang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Epigenetic Regulation and Intervention, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Zeqi Liu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Epigenetic Regulation and Intervention, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yiming Fan
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Hao-Tian Wang
- State Key Laboratory of Genetic Resources and Evolution/Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Zhuqiang Zhang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Epigenetic Regulation and Intervention, Chinese Academy of Sciences, Beijing, China
| | - Bing Zhu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
- Key Laboratory of Epigenetic Regulation and Intervention, Chinese Academy of Sciences, Beijing, China.
- New Cornerstone Science Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
32
|
Alcaráz N, Salcedo-Tello P, González-Barrios R, Torres-Arciga K, Guzmán-Ramos K. Underlying Mechanisms of the Protective Effects of Lifestyle Factors On Age-Related Diseases. Arch Med Res 2024; 55:103014. [PMID: 38861840 DOI: 10.1016/j.arcmed.2024.103014] [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: 12/07/2023] [Revised: 05/15/2024] [Accepted: 05/30/2024] [Indexed: 06/13/2024]
Abstract
The rise in life expectancy has significantly increased the occurrence of age-related chronic diseases, leading to escalating expenses for both society and individuals. Among the main factors influencing health and lifespan, lifestyle takes a forefront position. Specifically, nutrition, mental activity, and physical exercise influence the molecular and functional mechanisms that contribute to the prevention of major age-related diseases. Gaining deeper insights into the mechanisms that drive the positive effects of healthy lifestyles is valuable for creating interventions to prevent or postpone the development of chronic degenerative diseases. This review summarizes the main mechanisms that underlie the positive effect of lifestyle factors in counteracting the major age-related diseases involving brain health, musculoskeletal function, cancer, frailty, and cardiovascular diseases, among others. This knowledge will help to identify high-risk populations for targeted intervention trials and discover new biomarkers associated with healthy aging.
Collapse
Affiliation(s)
- Nicolás Alcaráz
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pamela Salcedo-Tello
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Rodrigo González-Barrios
- Instituto Nacional de Cancerología, Laboratorio de regulación de la cromatina y genómica, Mexico City, México
| | - Karla Torres-Arciga
- Instituto Nacional de Cancerología, Laboratorio de regulación de la cromatina y genómica, Mexico City, México; Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Kioko Guzmán-Ramos
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana, Unidad Lerma, Mexico State, Mexico.
| |
Collapse
|
33
|
McInvale JJ, Canoll P, Hargus G. Induced pluripotent stem cell models as a tool to investigate and test fluid biomarkers in Alzheimer's disease and frontotemporal dementia. Brain Pathol 2024; 34:e13231. [PMID: 38246596 PMCID: PMC11189780 DOI: 10.1111/bpa.13231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 11/29/2023] [Indexed: 01/23/2024] Open
Abstract
Neurodegenerative diseases are increasing in prevalence and comprise a large socioeconomic burden on patients and their caretakers. The need for effective therapies and avenues for disease prevention and monitoring is of paramount importance. Fluid biomarkers for neurodegenerative diseases have gained a variety of uses, including informing participant selection for clinical trials, lending confidence to clinical diagnosis and disease staging, determining prognosis, and monitoring therapeutic response. Their role is expected to grow as disease-modifying therapies start to be available to a broader range of patients and as prevention strategies become established. Many of the underlying molecular mechanisms of currently used biomarkers are incompletely understood. Animal models and in vitro systems using cell lines have been extensively employed but face important translatability limitations. Induced pluripotent stem cell (iPSC) technology, where a theoretically unlimited range of cell types can be reprogrammed from peripheral cells sampled from patients or healthy individuals, has gained prominence over the last decade. It is a promising avenue to study physiological and pathological biomarker function and response to experimental therapeutics. Such systems are amenable to high-throughput drug screening or multiomics readouts such as transcriptomics, lipidomics, and proteomics for biomarker discovery, investigation, and validation. The present review describes the current state of biomarkers in the clinical context of neurodegenerative diseases, with a focus on Alzheimer's disease and frontotemporal dementia. We include a discussion of how iPSC models have been used to investigate and test biomarkers such as amyloid-β, phosphorylated tau, neurofilament light chain or complement proteins, and even nominate novel biomarkers. We discuss the limitations of current iPSC methods, mentioning alternatives such as coculture systems and three-dimensional organoids which address some of these concerns. Finally, we propose exciting prospects for stem cell transplantation paradigms using animal models as a preclinical tool to study biomarkers in the in vivo context.
Collapse
Affiliation(s)
- Julie J. McInvale
- Department of Pathology and Cell BiologyColumbia UniversityNew YorkNew YorkUSA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia UniversityNew YorkNew YorkUSA
- Medical Scientist Training Program, Columbia UniversityNew YorkNew YorkUSA
| | - Peter Canoll
- Department of Pathology and Cell BiologyColumbia UniversityNew YorkNew YorkUSA
| | - Gunnar Hargus
- Department of Pathology and Cell BiologyColumbia UniversityNew YorkNew YorkUSA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia UniversityNew YorkNew YorkUSA
| |
Collapse
|
34
|
Zhuang X, Wang Q, Joost S, Ferrena A, Humphreys DT, Li Z, Blum M, Bastl K, Ding S, Landais Y, Zhan Y, Zhao Y, Chaligne R, Lee JH, Carrasco SE, Bhanot UK, Koche RP, Bott MJ, Katajisto P, Soto-Feliciano YM, Pisanic T, Thomas T, Zheng D, Wong ES, Tammela T. Aging limits stemness and tumorigenesis in the lung by reprogramming iron homeostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.23.600305. [PMID: 38979280 PMCID: PMC11230188 DOI: 10.1101/2024.06.23.600305] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Aging is associated with a decline in the number and fitness of adult stem cells 1-4 . Aging-associated loss of stemness is posited to suppress tumorigenesis 5,6 , but this hypothesis has not been tested in vivo . Here, using physiologically aged autochthonous genetically engineered mouse models and primary cells 7,8 , we demonstrate aging suppresses lung cancer initiation and progression by degrading stemness of the alveolar cell of origin. This phenotype is underpinned by aging-associated induction of the transcription factor NUPR1 and its downstream target lipocalin-2 in the cell of origin in mice and humans, leading to a functional iron insufficiency in the aged cells. Genetic inactivation of the NUPR1-lipocalin-2 axis or iron supplementation rescue stemness and promote tumorigenic potential of aged alveolar cells. Conversely, targeting the NUPR1- lipocalin-2 axis is detrimental to young alveolar cells via induction of ferroptosis. We find that aging-associated DNA hypomethylation at specific enhancer sites associates with elevated NUPR1 expression, which is recapitulated in young alveolar cells by inhibition of DNA methylation. We uncover that aging drives a functional iron insufficiency, which leads to loss of stemness and tumorigenesis, but promotes resistance to ferroptosis. These findings have significant implications for the therapeutic modulation of cellular iron homeostasis in regenerative medicine and in cancer prevention. Furthermore, our findings are consistent with a model whereby most human cancers initiate in young individuals, revealing a critical window for such cancer prevention efforts.
Collapse
|
35
|
Zhang S, Xiao X, Yi Y, Wang X, Zhu L, Shen Y, Lin D, Wu C. Tumor initiation and early tumorigenesis: molecular mechanisms and interventional targets. Signal Transduct Target Ther 2024; 9:149. [PMID: 38890350 PMCID: PMC11189549 DOI: 10.1038/s41392-024-01848-7] [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: 01/01/2024] [Revised: 04/23/2024] [Accepted: 04/27/2024] [Indexed: 06/20/2024] Open
Abstract
Tumorigenesis is a multistep process, with oncogenic mutations in a normal cell conferring clonal advantage as the initial event. However, despite pervasive somatic mutations and clonal expansion in normal tissues, their transformation into cancer remains a rare event, indicating the presence of additional driver events for progression to an irreversible, highly heterogeneous, and invasive lesion. Recently, researchers are emphasizing the mechanisms of environmental tumor risk factors and epigenetic alterations that are profoundly influencing early clonal expansion and malignant evolution, independently of inducing mutations. Additionally, clonal evolution in tumorigenesis reflects a multifaceted interplay between cell-intrinsic identities and various cell-extrinsic factors that exert selective pressures to either restrain uncontrolled proliferation or allow specific clones to progress into tumors. However, the mechanisms by which driver events induce both intrinsic cellular competency and remodel environmental stress to facilitate malignant transformation are not fully understood. In this review, we summarize the genetic, epigenetic, and external driver events, and their effects on the co-evolution of the transformed cells and their ecosystem during tumor initiation and early malignant evolution. A deeper understanding of the earliest molecular events holds promise for translational applications, predicting individuals at high-risk of tumor and developing strategies to intercept malignant transformation.
Collapse
Affiliation(s)
- Shaosen Zhang
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Xinyi Xiao
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Yonglin Yi
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Xinyu Wang
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Lingxuan Zhu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
- Changping Laboratory, 100021, Beijing, China
| | - Yanrong Shen
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Dongxin Lin
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
- Changping Laboratory, 100021, Beijing, China.
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, China.
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou, 510060, China.
| | - Chen Wu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
- Key Laboratory of Cancer Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
- Changping Laboratory, 100021, Beijing, China.
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, China.
- CAMS Oxford Institute, Chinese Academy of Medical Sciences, 100006, Beijing, China.
| |
Collapse
|
36
|
Li CZ, Haghani A, Yan Q, Lu AT, Zhang J, Fei Z, Ernst J, Yang XW, Gladyshev VN, Robeck TR, Chavez AS, Cook JA, Dunnum JL, Raj K, Seluanov A, Gorbunova V, Horvath S. Epigenetic predictors of species maximum life span and other life-history traits in mammals. SCIENCE ADVANCES 2024; 10:eadm7273. [PMID: 38848365 PMCID: PMC11160467 DOI: 10.1126/sciadv.adm7273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 05/03/2024] [Indexed: 06/09/2024]
Abstract
By analyzing 15,000 samples from 348 mammalian species, we derive DNA methylation (DNAm) predictors of maximum life span (R = 0.89), gestation time (R = 0.96), and age at sexual maturity (R = 0.85). Our maximum life-span predictor indicates a potential innate longevity advantage for females over males in 17 mammalian species including humans. The DNAm maximum life-span predictions are not affected by caloric restriction or partial reprogramming. Genetic disruptions in the somatotropic axis such as growth hormone receptors have an impact on DNAm maximum life span only in select tissues. Cancer mortality rates show no correlation with our epigenetic estimates of life-history traits. The DNAm maximum life-span predictor does not detect variation in life span between individuals of the same species, such as between the breeds of dogs. Maximum life span is determined in part by an epigenetic signature that is an intrinsic species property and is distinct from the signatures that relate to individual mortality risk.
Collapse
Affiliation(s)
- Caesar Z. Li
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA, USA
- Johnson & Johnson Innovative Medicine, Spring House, PA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Amin Haghani
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Altos Labs, San Diego, CA, USA
| | - Qi Yan
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA, USA
- Altos Labs, San Diego, CA, USA
| | - Ake T. Lu
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Altos Labs, San Diego, CA, USA
| | - Joshua Zhang
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zhe Fei
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Statistics, University of California, Riverside, Riverside, CA, USA
| | - Jason Ernst
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - X. William Yang
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Vadim N. Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Todd R. Robeck
- Zoological Operations, SeaWorld Parks and Entertainment Inc., Orlando, FL, USA
| | - Andreas S. Chavez
- Department of Evolution, Ecology and Organismal Biology, The Ohio State University, Columbus, OH, USA
- Translational Data Analytics Institute, The Ohio State University, Columbus, OH, USA
| | - Joseph A. Cook
- Department of Biology and Museum of Southwestern Biology, University of New Mexico, Albuquerque, NM, USA
| | - Jonathan L. Dunnum
- Department of Biology and Museum of Southwestern Biology, University of New Mexico, Albuquerque, NM, USA
| | | | - Andrei Seluanov
- Departments of Biology and Medicine, University of Rochester, Rochester, NY, USA
| | - Vera Gorbunova
- Departments of Biology and Medicine, University of Rochester, Rochester, NY, USA
| | - Steve Horvath
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Altos Labs, San Diego, CA, USA
- Altos Labs, Cambridge, UK
| |
Collapse
|
37
|
Shrestha B, Nieminen AI, Matilainen O. Loss of the histone chaperone UNC-85/ASF1 inhibits the epigenome-mediated longevity and modulates the activity of one-carbon metabolism. Cell Stress Chaperones 2024; 29:392-403. [PMID: 38608859 PMCID: PMC11039323 DOI: 10.1016/j.cstres.2024.04.003] [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: 01/22/2024] [Revised: 04/05/2024] [Accepted: 04/08/2024] [Indexed: 04/14/2024] Open
Abstract
Histone H3/H4 chaperone anti-silencing function 1 (ASF1) is a conserved factor mediating nucleosomal assembly and disassembly, playing crucial roles in processes such as replication, transcription, and DNA repair. Nevertheless, its involvement in aging has remained unclear. Here, we utilized the model organism Caenorhabditis elegans to demonstrate that the loss of UNC-85, the homolog of ASF1, leads to a shortened lifespan in a multicellular organism. Furthermore, we show that UNC-85 is required for epigenome-mediated longevity, as knockdown of the histone H3 lysine K4 methyltransferase ash-2 does not extend the lifespan of unc-85 mutants. In this context, we found that the longevity-promoting ash-2 RNA interference enhances UNC-85 activity by increasing its nuclear localization. Finally, our data indicate that the loss of UNC-85 increases the activity of one-carbon metabolism, and that downregulation of the one-carbon metabolism component dao-3/MTHFD2 partially rescues the short lifespan of unc-85 mutants. Together, these findings reveal UNC-85/ASF1 as a modulator of the central metabolic pathway and a factor regulating a pro-longevity response, thus shedding light on a mechanism of how nucleosomal maintenance associates with aging.
Collapse
Affiliation(s)
- Bideep Shrestha
- The Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Anni I Nieminen
- FIMM Metabolomics Unit, Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Olli Matilainen
- The Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
38
|
Tong H, Dwaraka VB, Chen Q, Luo Q, Lasky-Su JA, Smith R, Teschendorff AE. Quantifying the stochastic component of epigenetic aging. NATURE AGING 2024; 4:886-901. [PMID: 38724732 PMCID: PMC11186785 DOI: 10.1038/s43587-024-00600-8] [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: 09/22/2023] [Accepted: 02/21/2024] [Indexed: 05/15/2024]
Abstract
DNA methylation clocks can accurately estimate chronological age and, to some extent, also biological age, yet the process by which age-associated DNA methylation (DNAm) changes are acquired appears to be quasi-stochastic, raising a fundamental question: how much of an epigenetic clock's predictive accuracy could be explained by a stochastic process of DNAm change? Here, using DNAm data from sorted immune cells, we build realistic simulation models, subsequently demonstrating in over 22,770 sorted and whole-blood samples from 25 independent cohorts that approximately 66-75% of the accuracy underpinning Horvath's clock could be driven by a stochastic process. This fraction increases to 90% for the more accurate Zhang's clock, but is lower (63%) for the PhenoAge clock, suggesting that biological aging is reflected by nonstochastic processes. Confirming this, we demonstrate that Horvath's age acceleration in males and PhenoAge's age acceleration in severe coronavirus disease 2019 cases and smokers are not driven by an increased rate of stochastic change but by nonstochastic processes. These results significantly deepen our understanding and interpretation of epigenetic clocks.
Collapse
Affiliation(s)
- Huige Tong
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | | | - Qingwen Chen
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Qi Luo
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jessica A Lasky-Su
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Andrew E Teschendorff
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
39
|
Missong H, Joshi R, Khullar N, Thareja S, Navik U, Bhatti GK, Bhatti JS. Nutrient-epigenome interactions: Implications for personalized nutrition against aging-associated diseases. J Nutr Biochem 2024; 127:109592. [PMID: 38325612 DOI: 10.1016/j.jnutbio.2024.109592] [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: 10/15/2023] [Revised: 01/28/2024] [Accepted: 01/30/2024] [Indexed: 02/09/2024]
Abstract
Aging is a multifaceted process involving genetic and environmental interactions often resulting in epigenetic changes, potentially leading to aging-related diseases. Various strategies, like dietary interventions and calorie restrictions, have been employed to modify these epigenetic landscapes. A burgeoning field of interest focuses on the role of microbiota in human health, emphasizing system biology and computational approaches. These methods help decipher the intricate interplay between diet and gut microbiota, facilitating the creation of personalized nutrition strategies. In this review, we analysed the mechanisms related to nutritional interventions while highlighting the influence of dietary strategies, like calorie restriction and intermittent fasting, on microbial composition and function. We explore how gut microbiota affects the efficacy of interventions using tools like multi-omics data integration, network analysis, and machine learning. These tools enable us to pinpoint critical regulatory elements and generate individualized models for dietary responses. Lastly, we emphasize the need for a deeper comprehension of nutrient-epigenome interactions and the potential of personalized nutrition informed by individual genetic and epigenetic profiles. As knowledge and technology advance, dietary epigenetics stands on the cusp of reshaping our strategy against aging and related diseases.
Collapse
Affiliation(s)
- Hemi Missong
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Riya Joshi
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Naina Khullar
- Department of Zoology, Mata Gujri College, Fatehgarh Sahib, Punjab, India
| | - Suresh Thareja
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, Punjab, India
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Bathinda, Punjab, India
| | - Gurjit Kaur Bhatti
- Department of Medical Lab Technology, University Institute of Applied Health Sciences, Chandigarh University, Mohali, Punjab, India.
| | - Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, India.
| |
Collapse
|
40
|
Lv L, Chen Q, Lu J, Zhao Q, Wang H, Li J, Yuan K, Dong Z. Potential regulatory role of epigenetic modifications in aging-related heart failure. Int J Cardiol 2024; 401:131858. [PMID: 38360101 DOI: 10.1016/j.ijcard.2024.131858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 12/27/2023] [Accepted: 02/10/2024] [Indexed: 02/17/2024]
Abstract
Heart failure (HF) is a serious clinical syndrome and a serious development or advanced stage of various heart diseases. Aging is an independent factor that causes pathological damage in cardiomyopathy and participates in the occurrence of HF at the molecular level by affecting mechanisms such as telomere shortening and mitochondrial dysfunction. Epigenetic changes have a significant impact on the aging process, and there is increasing evidence that genetic and epigenetic changes are key features of aging and aging-related diseases. Epigenetic modifications can affect genetic information by changing the chromatin state without changing the DNA sequence. Most of the genetic loci that are highly associated with cardiovascular diseases (CVD) are located in non-coding regions of the genome; therefore, the epigenetic mechanism of CVD has attracted much attention. In this review, we focus on the molecular mechanisms of HF during aging and epigenetic modifications mediating aging-related HF, emphasizing that epigenetic mechanisms play an important role in the pathogenesis of aging-related CVD and can be used as potential diagnostic and prognostic biomarkers, as well as therapeutic targets.
Collapse
Affiliation(s)
- Lin Lv
- Department of Pharmacy, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - QiuYu Chen
- The Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jing Lu
- Department of Pharmacy, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qi Zhao
- Department of Pharmacy, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - HongYan Wang
- Department of Pharmacy, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - JiaHao Li
- Department of Pharmacy, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - KeYing Yuan
- Department of Pharmacy, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - ZengXiang Dong
- The Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, First Affiliated Hospital of Harbin Medical University, Harbin, China; NHC Key Laboratory of Cell Transplantation, First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
41
|
Morandini F, Rechsteiner C, Perez K, Praz V, Lopez Garcia G, Hinte LC, von Meyenn F, Ocampo A. ATAC-clock: An aging clock based on chromatin accessibility. GeroScience 2024; 46:1789-1806. [PMID: 37924441 PMCID: PMC10828344 DOI: 10.1007/s11357-023-00986-0] [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: 07/15/2023] [Accepted: 10/14/2023] [Indexed: 11/06/2023] Open
Abstract
The establishment of aging clocks highlighted the strong link between changes in DNA methylation and aging. Yet, it is not known if other epigenetic features could be used to predict age accurately. Furthermore, previous studies have observed a lack of effect of age-related changes in DNA methylation on gene expression, putting the interpretability of DNA methylation-based aging clocks into question. In this study, we explore the use of chromatin accessibility to construct aging clocks. We collected blood from 159 human donors and generated chromatin accessibility, transcriptomic, and cell composition data. We investigated how chromatin accessibility changes during aging and constructed a novel aging clock with a median absolute error of 5.27 years. The changes in chromatin accessibility used by the clock were strongly related to transcriptomic alterations, aiding clock interpretation. We additionally show that our chromatin accessibility clock performs significantly better than a transcriptomic clock trained on matched samples. In conclusion, we demonstrate that the clock relies on cell-intrinsic chromatin accessibility alterations rather than changes in cell composition. Further, we present a new approach to construct epigenetic aging clocks based on chromatin accessibility, which bear a direct link to age-related transcriptional alterations, but which allow for more accurate age predictions than transcriptomic clocks.
Collapse
Affiliation(s)
- Francesco Morandini
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Cheyenne Rechsteiner
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Kevin Perez
- EPITERNA SA, Route de la Corniche 5, Epalinges, Switzerland
| | - Viviane Praz
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Guillermo Lopez Garcia
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
- Departamento de Lenguajes y Ciencias de la Computación, Universidad de Málaga, Málaga, Spain
| | - Laura C Hinte
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | | | - Alejandro Ocampo
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland.
- EPITERNA SA, Route de la Corniche 5, Epalinges, Switzerland.
| |
Collapse
|
42
|
Soheili-Nezhad S, Ibáñez-Solé O, Izeta A, Hoeijmakers JHJ, Stoeger T. Time is ticking faster for long genes in aging. Trends Genet 2024; 40:299-312. [PMID: 38519330 PMCID: PMC11003850 DOI: 10.1016/j.tig.2024.01.009] [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: 11/21/2023] [Revised: 01/30/2024] [Accepted: 01/30/2024] [Indexed: 03/24/2024]
Abstract
Recent studies of aging organisms have identified a systematic phenomenon, characterized by a negative correlation between gene length and their expression in various cell types, species, and diseases. We term this phenomenon gene-length-dependent transcription decline (GLTD) and suggest that it may represent a bottleneck in the transcription machinery and thereby significantly contribute to aging as an etiological factor. We review potential links between GLTD and key aging processes such as DNA damage and explore their potential in identifying disease modification targets. Notably, in Alzheimer's disease, GLTD spotlights extremely long synaptic genes at chromosomal fragile sites (CFSs) and their vulnerability to postmitotic DNA damage. We suggest that GLTD is an integral element of biological aging.
Collapse
Affiliation(s)
- Sourena Soheili-Nezhad
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands; Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Olga Ibáñez-Solé
- Stem Cells & Aging Group, Biogipuzkoa Health Research Institute, Donostia-San Sebastián, Spain; Institute for Genome Stability in Aging and Disease, Medical Faculty, University and University Hospital of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany; Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), Center for Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany
| | - Ander Izeta
- Stem Cells & Aging Group, Biogipuzkoa Health Research Institute, Donostia-San Sebastián, Spain; Tecnun-University of Navarra, 20018 Donostia-San Sebastian, Spain.
| | - Jan H J Hoeijmakers
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands; University of Cologne, Faculty of Medicine, Cluster of Excellence for Aging Research, Institute for Genome Stability in Ageing and Disease, Cologne, Germany; Princess Maxima Center for Pediatric Oncology, Oncode Institute, Utrecht, The Netherlands.
| | - Thomas Stoeger
- Feinberg School of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, IL, USA; Potocsnak Longevity Institute, Northwestern University, Chicago, IL, USA; Simpson Querrey Lung Institute for Translational Science, Chicago, IL, USA.
| |
Collapse
|
43
|
Mahdi I, Imbimbo P, Annaz H, Bakrim WB, Sahri N, Alaoui A, Monti DM, Sobeh M. Profiling of Petroselinum sativum (mill.) fuss phytoconstituents and assessment of their biocompatibility, antioxidant, anti-aging, wound healing, and antibacterial activities. Front Nutr 2024; 11:1338482. [PMID: 38505264 PMCID: PMC10948610 DOI: 10.3389/fnut.2024.1338482] [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: 11/14/2023] [Accepted: 02/21/2024] [Indexed: 03/21/2024] Open
Abstract
Petroselinum sativum, known as parsley, is a fragrant herb that possesses a rich heritage of utilization in traditional medicinal practices. In this study, we annotated the phytocontents of the aqueous and ethanolic extracts of P. sativum and investigated their antioxidant, cytoprotective, antiaging, wound healing, and antibacterial activities. LC-MS/MS analysis of both extracts revealed the presence of 47 compounds belonging to diverse groups including organic acids, phenolic acids, and flavonoids. By MTT assay, the extracts were fully biocompatible on immortalized human keratinocytes (HaCaT) while they inhibited intracellular ROS formation (DCFDA assay) and prevented GSH depletion (DTNB assay) upon UVA exposure. In addition, the extracts were potent in inhibiting the in vitro activities of skin-related enzymes mainly elastase, tyrosinase, collagenase and hyaluronidase. Using the scratch assay, P. sativum aqueous extract significantly enhanced wound closure when compared to untreated HaCaT cells. Moreover, both extracts inhibited Pseudomonas aeruginosa's growth, reduced biofilm formation, and impaired the swimming and swarming motilities. Also, the aqueous extract was able to inhibit the production of bacterial pigments on plates. These findings strongly suggest the usefulness of P. sativum as a source of phytochemicals suitable for dermo-cosmeceutical applications.
Collapse
Affiliation(s)
- Ismail Mahdi
- AgroBioSciences Program, College of Agriculture and Environmental Sciences, University Mohammed VI Polytechnic, Ben Guerir, Morocco
| | - Paola Imbimbo
- Department of Chemical Sciences, University of Napoli Federico II, Complesso Universitario Monte Sant’Angelo, Napoli, Italy
| | - Hassan Annaz
- AgroBioSciences Program, College of Agriculture and Environmental Sciences, University Mohammed VI Polytechnic, Ben Guerir, Morocco
| | - Widad Ben Bakrim
- AgroBioSciences Program, College of Agriculture and Environmental Sciences, University Mohammed VI Polytechnic, Ben Guerir, Morocco
| | - Nihad Sahri
- AgroBioSciences Program, College of Agriculture and Environmental Sciences, University Mohammed VI Polytechnic, Ben Guerir, Morocco
- Physio-Chemical Laboratory of Inorganic and Organic Materials (LPCMIO), Materials Science Center (MSC), Ecole Normale Supérieure, Mohammed V University, Rabat, Morocco
| | - Asmae Alaoui
- Physio-Chemical Laboratory of Inorganic and Organic Materials (LPCMIO), Materials Science Center (MSC), Ecole Normale Supérieure, Mohammed V University, Rabat, Morocco
| | - Daria Maria Monti
- Department of Chemical Sciences, University of Napoli Federico II, Complesso Universitario Monte Sant’Angelo, Napoli, Italy
| | - Mansour Sobeh
- AgroBioSciences Program, College of Agriculture and Environmental Sciences, University Mohammed VI Polytechnic, Ben Guerir, Morocco
| |
Collapse
|
44
|
Falckenhayn C, Bienkowska A, Söhle J, Wegner K, Raddatz G, Kristof B, Kuck D, Siegner R, Kaufmann R, Korn J, Baumann S, Lange D, Schepky A, Völzke H, Kaderali L, Winnefeld M, Lyko F, Grönniger E. Identification of dihydromyricetin as a natural DNA methylation inhibitor with rejuvenating activity in human skin. FRONTIERS IN AGING 2024; 4:1258184. [PMID: 38500495 PMCID: PMC10944877 DOI: 10.3389/fragi.2023.1258184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 12/12/2023] [Indexed: 03/20/2024]
Abstract
Changes in DNA methylation patterning have been reported to be a key hallmark of aged human skin. The altered DNA methylation patterns are correlated with deregulated gene expression and impaired tissue functionality, leading to the well-known skin aging phenotype. Searching for small molecules, which correct the aged methylation pattern therefore represents a novel and attractive strategy for the identification of anti-aging compounds. DNMT1 maintains epigenetic information by copying methylation patterns from the parental (methylated) strand to the newly synthesized strand after DNA replication. We hypothesized that a modest inhibition of this process promotes the restoration of the ground-state epigenetic pattern, thereby inducing rejuvenating effects. In this study, we screened a library of 1800 natural substances and 640 FDA-approved drugs and identified the well-known antioxidant and anti-inflammatory molecule dihydromyricetin (DHM) as an inhibitor of the DNA methyltransferase DNMT1. DHM is the active ingredient of several plants with medicinal use and showed robust inhibition of DNMT1 in biochemical assays. We also analyzed the effect of DHM in cultivated keratinocytes by array-based methylation profiling and observed a moderate, but significant global hypomethylation effect upon treatment. To further characterize DHM-induced methylation changes, we used published DNA methylation clocks and newly established age predictors to demonstrate that the DHM-induced methylation change is associated with a reduction in the biological age of the cells. Further studies also revealed re-activation of age-dependently hypermethylated and silenced genes in vivo and a reduction in age-dependent epidermal thinning in a 3-dimensional skin model. Our findings thus establish DHM as an epigenetic inhibitor with rejuvenating effects for aged human skin.
Collapse
Affiliation(s)
| | - Agata Bienkowska
- Beiersdorf AG, Research and Development, Hamburg, Germany
- Institute for Bioinformatics, University Medicine Greifswald, Greifswald, Germany
| | - Jörn Söhle
- Beiersdorf AG, Research and Development, Hamburg, Germany
| | - Katrin Wegner
- Beiersdorf AG, Research and Development, Hamburg, Germany
| | - Guenter Raddatz
- Division of Epigenetics, DKFZ-ZMBH Alliance, German Cancer Research Center, Heidelberg, Germany
| | - Boris Kristof
- Beiersdorf AG, Research and Development, Hamburg, Germany
| | - Dirk Kuck
- Division of Epigenetics, DKFZ-ZMBH Alliance, German Cancer Research Center, Heidelberg, Germany
| | - Ralf Siegner
- Beiersdorf AG, Research and Development, Hamburg, Germany
| | - Ronny Kaufmann
- Beiersdorf AG, Research and Development, Hamburg, Germany
| | - Julia Korn
- Beiersdorf AG, Research and Development, Hamburg, Germany
| | - Sascha Baumann
- Beiersdorf AG, Research and Development, Hamburg, Germany
| | - Daniela Lange
- Beiersdorf AG, Research and Development, Hamburg, Germany
| | | | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Lars Kaderali
- Institute for Bioinformatics, University Medicine Greifswald, Greifswald, Germany
| | - Marc Winnefeld
- Beiersdorf AG, Research and Development, Hamburg, Germany
| | - Frank Lyko
- Division of Epigenetics, DKFZ-ZMBH Alliance, German Cancer Research Center, Heidelberg, Germany
| | - Elke Grönniger
- Beiersdorf AG, Research and Development, Hamburg, Germany
| |
Collapse
|
45
|
Theodore Armand TP, Kim HC, Kim JI. Digital Anti-Aging Healthcare: An Overview of the Applications of Digital Technologies in Diet Management. J Pers Med 2024; 14:254. [PMID: 38540996 PMCID: PMC10970731 DOI: 10.3390/jpm14030254] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 02/21/2024] [Accepted: 02/25/2024] [Indexed: 04/30/2025] Open
Abstract
Diet management has long been an important practice in healthcare, enabling individuals to get an insight into their nutrient intake, prevent diseases, and stay healthy. Traditional methods based on self-reporting, food diaries, and periodic assessments have been used for a long time to control dietary habits. These methods have shown limitations in accuracy, compliance, and real-time analysis. The rapid advancement of digital technologies has revolutionized healthcare, including the diet control landscape, allowing for innovative solutions to control dietary patterns and generate accurate and personalized recommendations. This study examines the potential of digital technologies in diet management and their effectiveness in anti-aging healthcare. After underlining the importance of nutrition in the aging process, we explored the applications of mobile apps, web-based platforms, wearables devices, sensors, the Internet of Things, artificial intelligence, blockchain, and other technologies in managing dietary patterns and improving health outcomes. The research further examines the effects of digital dietary control on anti-aging healthcare, including improved nutritional monitoring, personalized recommendations, and behavioral and sustainable changes in habits, leading to an expansion of longevity and health span. The challenges and limitations of digital diet monitoring are discussed, and some future directions are provided. Although many digital tools are used in diet control, their accuracy, effectiveness, and impact on health outcomes are not discussed much. This review consolidates the existing literature on digital diet management using emerging digital technologies to analyze their practical implications, guiding researchers, healthcare professionals, and policy makers toward personalized dietary management and healthy aging.
Collapse
Affiliation(s)
- Tagne Poupi Theodore Armand
- Institute of Digital Anti-Aging Healthcare, Inje University, Gimhae 50834, Republic of Korea; (T.P.T.A.); (H.-C.K.)
| | - Hee-Cheol Kim
- Institute of Digital Anti-Aging Healthcare, Inje University, Gimhae 50834, Republic of Korea; (T.P.T.A.); (H.-C.K.)
- College of AI Convergence, u-AHRC, Inje University, Gimhae 50834, Republic of Korea
| | - Jung-In Kim
- Institute of Digital Anti-Aging Healthcare, Inje University, Gimhae 50834, Republic of Korea; (T.P.T.A.); (H.-C.K.)
| |
Collapse
|
46
|
Dubey SK, Dubey R, Kleinman ME. Unraveling Histone Loss in Aging and Senescence. Cells 2024; 13:320. [PMID: 38391933 PMCID: PMC10886805 DOI: 10.3390/cells13040320] [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: 12/31/2023] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/24/2024] Open
Abstract
As the global population experiences a notable surge in aging demographics, the need to understand the intricate molecular pathways exacerbated by age-related stresses, including epigenetic dysregulation, becomes a priority. Epigenetic mechanisms play a critical role in driving age-related diseases through altered gene expression, genomic instability, and irregular chromatin remodeling. In this review, we focus on histones, a central component of the epigenome, and consolidate the key findings of histone loss and genome-wide redistribution as fundamental processes contributing to aging and senescence. The review provides insights into novel histone expression profiles, nucleosome occupancy, disruptions in higher-order chromatin architecture, and the emergence of noncanonical histone variants in the aging cellular landscape. Furthermore, we explore the current state of our understanding of the molecular mechanisms of histone deficiency in aging cells. Specific emphasis is placed on highlighting histone degradation pathways in the cell and studies that have explored potential strategies to mitigate histone loss or restore histone levels in aging cells. Finally, in addressing future perspectives, the insights gained from this review hold profound implications for advancing strategies that actively intervene in modulating histone expression profiles in the context of cellular aging and identifying potential therapeutic targets for alleviating a multitude of age-related diseases.
Collapse
Affiliation(s)
| | | | - Mark Ellsworth Kleinman
- Department of Surgery, East Tennessee State University, Johnson City, TN 37614, USA; (S.K.D.); (R.D.)
| |
Collapse
|
47
|
Yagi G, Qi H, Arai K, Kita YF, Kogi K, Morisaka T, Yoshioka M, Inoue-Murayama M. Non-invasive age estimation based on faecal DNA using methylation-sensitive high-resolution melting for Indo-Pacific bottlenose dolphins. Mol Ecol Resour 2024; 24:e13906. [PMID: 38041546 DOI: 10.1111/1755-0998.13906] [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: 07/09/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 12/03/2023]
Abstract
Age is necessary information for the study of life history of wild animals. A general method to estimate the age of odontocetes is counting dental growth layer groups (GLGs). However, this method is highly invasive as it requires the capture and handling of individuals to collect their teeth. Recently, the development of DNA-based age estimation methods has been actively studied as an alternative to such invasive methods, of which many have relied on used biopsy samples. However, if DNA-based age estimation can be developed from faecal samples, age estimation can be performed entirely non-invasively. We developed an age estimation model using the methylation rate of two gene regions, GRIA2 and CDKN2A, measured through methylation-sensitive high-resolution melting (MS-HRM) from faecal samples of wild Indo-Pacific bottlenose dolphins (Tursiops aduncus). The age of individuals was known through conducting longitudinal individual identification surveys underwater. Methylation rates were quantified from 36 samples collected from 30 individuals. Both gene regions showed a significant correlation between age and methylation rate. The age estimation model was constructed based on the methylation rates of both genes which achieved sufficient accuracy (after LOOCV: MAE = 5.08, R2 = 0.33) for the ecological studies of the Indo-Pacific bottlenose dolphins, with a lifespan of 40-50 years. This is the first study to report the use of non-invasive faecal samples to estimate the age of marine mammals.
Collapse
Affiliation(s)
- Genfu Yagi
- Graduate School of Bioresources, Mie University, Tsu, Mie, Japan
| | - Huiyuan Qi
- Wildlife Research Center, Kyoto University, Kyoto, Kyoto, Japan
| | - Kana Arai
- Wildlife Research Center, Kyoto University, Kyoto, Kyoto, Japan
| | - Yuki F Kita
- Department of Marine Biology and Sciences, School of Biological Sciences, Tokai University, Sapporo, Hokkaido, Japan
| | | | | | - Motoi Yoshioka
- Graduate School of Bioresources, Mie University, Tsu, Mie, Japan
| | | |
Collapse
|
48
|
Kordowitzki P, Graczyk S, Haghani A, Klutstein M. Oocyte Aging: A Multifactorial Phenomenon in A Unique Cell. Aging Dis 2024; 15:5-21. [PMID: 37307833 PMCID: PMC10796106 DOI: 10.14336/ad.2023.0527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 05/27/2023] [Indexed: 06/14/2023] Open
Abstract
The oocyte is considered to be the largest cell in mammalian species. Women hoping to become pregnant face a ticking biological clock. This is becoming increasingly challenging as an increase in life expectancy is accompanied by the tendency to conceive at older ages. With advancing maternal age, the fertilized egg will exhibit lower quality and developmental competence, which contributes to increased chances of miscarriage due to several causes such as aneuploidy, oxidative stress, epigenetics, or metabolic disorders. In particular, heterochromatin in oocytes and with it, the DNA methylation landscape undergoes changes. Further, obesity is a well-known and ever-increasing global problem as it is associated with several metabolic disorders. More importantly, both obesity and aging negatively affect female reproduction. However, among women, there is immense variability in age-related decline of oocytes' quantity, developmental competence, or quality. Herein, the relevance of obesity and DNA-methylation will be discussed as these aspects have a tremendous effect on female fertility, and it is a topic of continuous and widespread interest that has yet to be fully addressed for the mammalian oocyte.
Collapse
Affiliation(s)
- Pawel Kordowitzki
- Department of Preclinical and Basic Sciences, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Torun, Poland.
| | - Szymon Graczyk
- Department of Preclinical and Basic Sciences, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Torun, Poland.
| | - Amin Haghani
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Altos Labs, San Diego, CA, USA.
| | - Michael Klutstein
- Institute of Biomedical and Oral Research, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
49
|
Linders AN, Dias IB, López Fernández T, Tocchetti CG, Bomer N, Van der Meer P. A review of the pathophysiological mechanisms of doxorubicin-induced cardiotoxicity and aging. NPJ AGING 2024; 10:9. [PMID: 38263284 PMCID: PMC10806194 DOI: 10.1038/s41514-024-00135-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/02/2024] [Indexed: 01/25/2024]
Abstract
The population of cancer survivors is rapidly increasing due to improving healthcare. However, cancer therapies often have long-term side effects. One example is cancer therapy-related cardiac dysfunction (CTRCD) caused by doxorubicin: up to 9% of the cancer patients treated with this drug develop heart failure at a later stage. In recent years, doxorubicin-induced cardiotoxicity has been associated with an accelerated aging phenotype and cellular senescence in the heart. In this review we explain the evidence of an accelerated aging phenotype in the doxorubicin-treated heart by comparing it to healthy aged hearts, and shed light on treatment strategies that are proposed in pre-clinical settings. We will discuss the accelerated aging phenotype and the impact it could have in the clinic and future research.
Collapse
Affiliation(s)
- Annet Nicole Linders
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, Groningen, The Netherlands
| | - Itamar Braga Dias
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, Groningen, The Netherlands
| | - Teresa López Fernández
- Division of Cardiology, Cardiac Imaging and Cardio-Oncology Unit, La Paz University Hospital, IdiPAZ Research Institute, Madrid, Spain
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences (DISMET), Federico II University, Naples, Italy
- Centre for Basic and Clinical Immunology Research (CISI), Federico II University, Naples, Italy
- Interdepartmental Centre of Clinical and Translational Sciences (CIRCET), Federico II University, Naples, Italy
- Interdepartmental Hypertension Research Centre (CIRIAPA), Federico II University, Naples, Italy
| | - Nils Bomer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, Groningen, The Netherlands
| | - Peter Van der Meer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, Groningen, The Netherlands.
| |
Collapse
|
50
|
Urman MA, John NS, Jung T, Lee C. Aging disrupts spatiotemporal regulation of germline stem cells and niche integrity. Biol Open 2024; 13:bio060261. [PMID: 38156664 PMCID: PMC10810562 DOI: 10.1242/bio.060261] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 12/13/2023] [Indexed: 01/03/2024] Open
Abstract
A major factor driving stem cell decline is stem cell niche aging, but its molecular mechanism remains elusive. We use the Caenorhabditis elegans distal tip cell (DTC), the mesenchymal niche that employs Notch signaling to regulate germline stem cells (GSCs), as an in vivo niche aging model and delineate the molecular details of the DTC/niche aging process. Here, we demonstrate that a drastic decrease in C. elegans germline fecundity, which begins even in early adulthood, is mainly due to an age-induced disruption in spatial regulation of Notch-dependent transcription in the germline combined with a moderate reduction in Notch transcription at both tissue and cellular levels. Consequently, the Notch-responsive GSC pool shifts from the distal end of the gonad to a more proximal region, disrupting the distal-to-proximal germline polarity. We find that this GSC pool shift is due to a dislocation of the DTC/niche nucleus, which is associated with age-induced changes in the structure and morphology of the DTC/niche. Our findings reveal a critical link between physiological changes in the aging niche, their consequences in stem cell regulation, and germline tissue functions.
Collapse
Affiliation(s)
- Michelle A. Urman
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, 12222, USA
- The RNA Institute, University at Albany, State University of New York, Albany, NY, 12222, USA
| | - Nimmy S. John
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, 12222, USA
- The RNA Institute, University at Albany, State University of New York, Albany, NY, 12222, USA
| | - Tyler Jung
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, 12222, USA
| | - ChangHwan Lee
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, 12222, USA
- The RNA Institute, University at Albany, State University of New York, Albany, NY, 12222, USA
| |
Collapse
|