Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 14.807
Filter
1.
Sci Signal ; 17(853): eads9401, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39255337

ABSTRACT

Cell type-specific gene signatures in an aged brain imply biological links to age-related disorders.


Subject(s)
Aging , Aging/genetics , Aging/metabolism , Humans , Animals , Brain/metabolism
2.
Genome Biol ; 25(1): 240, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39242518

ABSTRACT

BACKGROUND: During aging, the human methylome undergoes both differential and variable shifts, accompanied by increased entropy. The distinction between variably methylated positions (VMPs) and differentially methylated positions (DMPs), their contribution to epigenetic age, and the role of cell type heterogeneity remain unclear. RESULTS: We conduct a comprehensive analysis of > 32,000 human blood methylomes from 56 datasets (age range = 6-101 years). We find a significant proportion of the blood methylome that is differentially methylated with age (48% DMPs; FDR < 0.005) and variably methylated with age (37% VMPs; FDR < 0.005), with considerable overlap between the two groups (59% of DMPs are VMPs). Bivalent and Polycomb regions become increasingly methylated and divergent between individuals, while quiescent regions lose methylation more uniformly. Both chronological and biological clocks, but not pace-of-aging clocks, show a strong enrichment for CpGs undergoing both mean and variance changes during aging. The accumulation of DMPs shifting towards a methylation fraction of 50% drives the increase in entropy, smoothening the epigenetic landscape. However, approximately a quarter of DMPs exhibit anti-entropic effects, opposing this direction of change. While changes in cell type composition minimally affect DMPs, VMPs and entropy measurements are moderately sensitive to such alterations. CONCLUSION: This study represents the largest investigation to date of genome-wide DNA methylation changes and aging in a single tissue, providing valuable insights into primary molecular changes relevant to chronological and biological aging.


Subject(s)
Aging , DNA Methylation , Epigenesis, Genetic , Epigenome , Humans , Aging/genetics , Aging/blood , Aged , Adult , Adolescent , Aged, 80 and over , Middle Aged , Young Adult , Child , CpG Islands , Male , Female
3.
Mol Cell Endocrinol ; 592: 112348, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39218056

ABSTRACT

The developmental origins of healthy and disease (DOHaD) concept has demonstrated a higher rate of chronic diseases in the adult population of individuals whose mothers experienced severe maternal protein restriction (MPR). Using proteomic and in silico analyses, we investigated the lung proteomic profile of young and aged rats exposed to MPR during pregnancy and lactation. Our results demonstrated that MPR lead to structural and immune system pathways changes, and this outcome is coupled with a rise in the PI3k-AKT-mTOR signaling pathway, with increased MMP-2 activity, and CD8 expression in the early life, with long-term effects with aging. This led to the identification of commonly or inversely differentially expressed targets in early life and aging, revealing dysregulated pathways related to the immune system, stress, muscle contraction, tight junctions, and hemostasis. We identified three miRNAs (miR-378a-3p, miR-378a-5p, let-7a-5p) that regulate four proteins (ACTN4, PPIA, HSPA5, CALM1) as probable epigenetic lung marks generated by MPR. In conclusion, MPR impacts the lungs early in life, increasing the possibility of long-lasting negative outcomes for respiratory disorders in the offspring.


Subject(s)
Lung , MicroRNAs , Proteomics , Animals , Female , Lung/metabolism , Male , Proteomics/methods , Pregnancy , MicroRNAs/genetics , MicroRNAs/metabolism , Rats , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/genetics , Diet, Protein-Restricted , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/genetics , Longevity/genetics , Rats, Wistar , Proto-Oncogene Proteins c-akt/metabolism , Proteome/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Aging/metabolism , Aging/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 2/genetics
4.
Nat Commun ; 15(1): 7966, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39261515

ABSTRACT

Age is a risk factor for hematologic malignancies. Attributes of the aging hematopoietic system include increased myelopoiesis, impaired adaptive immunity, and a functional decline of the hematopoietic stem cells (HSCs) that maintain hematopoiesis. Changes in the composition of diverse HSC subsets have been suggested to be responsible for age-related alterations, however, the underlying regulatory mechanisms are incompletely understood in the context of HSC heterogeneity. In this study, we investigated how distinct HSC subsets, separated by CD49b, functionally and molecularly change their behavior with age. We demonstrate that the lineage differentiation of both lymphoid-biased and myeloid-biased HSC subsets progressively shifts to a higher myeloid cellular output during aging. In parallel, we show that HSCs selectively undergo age-dependent gene expression and gene regulatory changes in a progressive manner, which is initiated already in the juvenile stage. Overall, our studies suggest that aging intrinsically alters both cellular and molecular properties of HSCs.


Subject(s)
Aging , Hematopoietic Stem Cells , Mice, Inbred C57BL , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Animals , Aging/genetics , Aging/physiology , Mice , Cell Differentiation , Cell Lineage/genetics , Hematopoiesis/genetics , Myeloid Cells/metabolism , Myeloid Cells/cytology , Male , Gene Expression Regulation , Female
5.
Front Immunol ; 15: 1347770, 2024.
Article in English | MEDLINE | ID: mdl-39267750

ABSTRACT

Introduction: The connection between aging and cancer is complex. Previous research has highlighted the association between the aging process of lung adenocarcinoma (LUAD) cells and the immune response, yet there remains a gap in confirming this through single-cell data validation. Here, we aim to develop a novel aging-related prognostic model for LUAD, and verify the alterations in the genome and immune microenvironment linked to cellular senescence. Methods: We integrated a comprehensive collection of senescence genes from the GenAge and CellAge databases and employed the least absolute shrinkage and selection operator (LASSO) Cox analysis to construct and validate a novel prognostic model for LUAD. This model was then utilized to examine the relationship between aging, tumor somatic mutations, and immune cell infiltration. Additionally, we explored the heterogeneity of senescence and intercellular communication within the LUAD tumor microenvironment (TME) through single-cell transcriptomic data analysis. Results: By exploring the expression profiles of 586 cellular senescence-related genes in 428 LUAD patients, we constructed an aging-related genes (ARGs) risk model included 10 ARGs and validated it as an independent prognostic predictor for LUAD patients. Notably, patients with low aging scores (LAS group) exhibited better survival, lower tumor mutation burden (TMB), lower somatic mutation frequency, lower tumor proliferation rate, and an immune activated phenotype compared to patients with high aging scores (HAS group). While the HAS group was enriched in tumor cells and showed a lower infiltration of CD8-CCR7, CD8- CXCL13, CD8-GNLY, FCGR3A NK cells, XCL1 NK cells, plasma cell (PC) and other immune subsets. Furthermore, the SPP1 and TENASCIN pathways, associated with tumor immune escape and tumor progression, were also enriched in the HAS group. Additionally, our study also indicated that senescence levels were heterogeneous in the LUAD tumor microenvironment (TME), especially with tumor cells in the LAS group showing higher age scores compared to those in the HAS group. Conclusions: Collectively, our findings underscore that ARRS through ARGs serves as a robust biomarker for the prognosis in LUAD.


Subject(s)
Adenocarcinoma of Lung , Cellular Senescence , Lung Neoplasms , Tumor Microenvironment , Humans , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/mortality , Adenocarcinoma of Lung/pathology , Cellular Senescence/genetics , Cellular Senescence/immunology , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Prognosis , Biomarkers, Tumor/genetics , Mutation , Male , Female , Gene Expression Regulation, Neoplastic , Transcriptome , Middle Aged , Gene Expression Profiling , Aged , Aging/immunology , Aging/genetics
6.
J Ovarian Res ; 17(1): 185, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39272131

ABSTRACT

BACKGROUND: In-depth understanding of dynamic expression profiles of human granulosa cells (GCs) during follicular development will contribute to the diagnostic and targeted interventions for female infertility. However, genome-scale analysis of long non-coding ribonucleic acid (lncRNA) in GCs across diverse developmental stages is challenging. Meanwhile, further research is needed to determine how aberrant lncRNA expression participates in ovarian diseases. METHODS: Granulosa cell-related lncRNAs data spanning five follicular development stages were retrieved and filtered from the NCBI dataset (GSE107746). Stage-specific lncRNA expression patterns and mRNA-lncRNA co-expression networks were identified with bioinformatic approaches. Subsequently, the expression pattern of SNHG18 was detected in GCs during ovarian aging. And SNHG18 siRNA or overexpression plasmids were transfected to SVOG cells in examining the regulatory roles of SNHG18 in GC proliferation and apoptosis. Moreover, whether PKCɛ/SNHG18 signaling take part in GC glycolysis via ENO1 were verified in SVOG cells. RESULTS: We demonstrated that GC-related lncRNAs were specifically expressed across different developmental stages, and coordinated crucial biological functions like mitotic cell cycle and metabolic processes in the folliculogenesis. Thereafter, we noticed a strong correlation of PRKCE and SNHG18 expression in our analysis. With downregulated SNHG18 of GCs identified in the context of ovarian aging, SNHG18 knockdown could further induce cell apoptosis, retard cell proliferation and exacerbate DNA damage in SVOG cell. Moreover, downregulated PKCɛ/SNHG18 pathway interrupted the SVOG cell glycolysis by lowering the ENO1 expression. CONCLUSIONS: Altogether, our results revealed that folliculogenesis-related lncRNA SNHG18 participated in the pathogenesis of ovarian aging, which may provide novel biomarkers for ovarian function and new insights for the infertility treatment.


Subject(s)
Apoptosis , Glycolysis , Granulosa Cells , RNA, Long Noncoding , Female , Humans , Aging/genetics , Aging/metabolism , Apoptosis/genetics , Glycolysis/genetics , Granulosa Cells/metabolism , Ovary/metabolism , Ovary/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
7.
BMC Genomics ; 23(Suppl 4): 866, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39294566

ABSTRACT

BACKGROUND: Aging is a complex, heterogeneous process that has multiple causes. Knowledge on genomic, epigenomic and transcriptomic changes during the aging process shed light on understanding the aging mechanism. A recent breakthrough in biotechnology, single cell RNAseq, is revolutionizing aging study by providing gene expression profile of the entire transcriptome of individual cells. Many interesting information could be inferred from this new type of data with the help of novel computational methods. RESULTS: In this manuscript a novel statistical method, penalized Latent Dirichlet Allocation (pLDA), is applied to an aging mouse blood scRNA-seq data set. A pipeline is built for cell type and aging prediction. The sequence of models in the pipeline take scRNA-seq expression counts as input, preprocess the data using pLDA and predict the cell type and aging status. CONCLUSIONS: pLDA learns a dimension reduced representation of the expression profile. This representation allows identification of cell types and has predictability of the age of cells.


Subject(s)
Aging , Animals , Mice , Aging/genetics , Single-Cell Analysis/methods , Blood Cells/metabolism , Transcriptome , Gene Expression Profiling/methods , Computational Biology/methods , Algorithms
8.
Clin Epigenetics ; 16(1): 133, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39300457

ABSTRACT

BACKGROUND: Epigenetic age accelerations (EAAs) are a promising new avenue of research, yet their investigation in subacute thyroiditis (SAT) remains scarce. Our study endeavors to fill this void by exploring the potential causal association between EAAs and SAT. METHODS: Our study utilized publicly available genome-wide association study (GWAS) data of European ancestry to conduct a bidirectional Mendelian randomization (MR) study. Five MR methods were employed to measure causal association between EAAs and SAT multiple analyses were utilized to perform quality control. RESULTS: Our study evaluated causal association between SAT and four EAAs, included GrimAge acceleration (GrimAA), Hannum age acceleration (HannumAA), PhenoAge acceleration (PhenoAA), intrinsic epigenetic age acceleration (IEAA). Results showed that there is a significant causal association between PhenoAA and SAT (OR 1.109, 95% CI 1.000-1.228, p = 0.049, by IVW method). On the contrary, SAT was associated with IEAA (OR 0.933, 95% CI 0.884-0.984, p = 0.011, by IVW method; OR 0.938, 95% CI 0.881-0.998, p = 0.043, by weighted median method). Leave-one-out sensitivity analysis, heterogeneity test, pleiotropy test, and MR-PRESSO analysis provide good quality control. CONCLUSION: The bidirectional MR analysis concluded that an increase in PhenoAA was correlated with a higher risk of SAT, indicating a potential causal relationship between PhenoAA and risk of SAT. Conversely, SAT was found to be closely associated with IEAA, suggesting that SAT may accelerate the aging process. Slowing down biological aging has emerged as a new research direction in curbing SAT.


Subject(s)
Epigenesis, Genetic , Genome-Wide Association Study , Mendelian Randomization Analysis , Thyroiditis, Subacute , Humans , Mendelian Randomization Analysis/methods , Genome-Wide Association Study/methods , Epigenesis, Genetic/genetics , Thyroiditis, Subacute/genetics , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Female , DNA Methylation/genetics , Male , Risk Factors , Aging/genetics
10.
Nat Commun ; 15(1): 7858, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39251642

ABSTRACT

Clonal hematopoiesis of indeterminate potential (CHIP) is linked to diverse aging-related diseases, including hematologic malignancy and atherosclerotic cardiovascular disease (ASCVD). While CHIP is common among older adults, the underlying factors driving its development are largely unknown. To address this, we performed whole-exome sequencing on 8,374 blood DNA samples collected from 4,187 Atherosclerosis Risk in Communities Study (ARIC) participants over a median follow-up of 21 years. During this period, 735 participants developed incident CHIP. Splicing factor genes (SF3B1, SRSF2, U2AF1, and ZRSR2) and TET2 CHIP grow significantly faster than DNMT3A non-R882 clones. We find that age at baseline and sex significantly influence the incidence of CHIP, while ASCVD and other traditional ASCVD risk factors do not exhibit such associations. Additionally, baseline synonymous passenger mutations are strongly associated with CHIP status and are predictive of new CHIP clone acquisition and clonal growth over extended follow-up, providing valuable insights into clonal dynamics of aging hematopoietic stem and progenitor cells. This study also reveals associations between germline genetic variants and incident CHIP. Our comprehensive longitudinal assessment yields insights into cell-intrinsic and -extrinsic factors contributing to the development and progression of CHIP clones in older adults.


Subject(s)
Clonal Hematopoiesis , Dioxygenases , Humans , Clonal Hematopoiesis/genetics , Male , Female , Aged , Longitudinal Studies , Middle Aged , Dioxygenases/genetics , DNA Methyltransferase 3A , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , Atherosclerosis/genetics , Risk Factors , Exome Sequencing , DNA-Binding Proteins/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Aging/genetics , Incidence , Mutation
11.
Elife ; 132024 Sep 16.
Article in English | MEDLINE | ID: mdl-39283662

ABSTRACT

Over the past several decades, a trend toward delayed childbirth has led to increases in parental age at the time of conception. Sperm epigenome undergoes age-dependent changes increasing risks of adverse conditions in offspring conceived by fathers of advanced age. The mechanism(s) linking paternal age with epigenetic changes in sperm remain unknown. The sperm epigenome is shaped in a compartment protected by the blood-testes barrier (BTB) known to deteriorate with age. Permeability of the BTB is regulated by the balance of two mTOR complexes in Sertoli cells where mTOR complex 1 (mTORC1) promotes the opening of the BTB and mTOR complex 2 (mTORC2) promotes its integrity. We hypothesized that this balance is also responsible for age-dependent changes in the sperm epigenome. To test this hypothesis, we analyzed reproductive outcomes, including sperm DNA methylation in transgenic mice with Sertoli cell-specific suppression of mTORC1 (Rptor KO) or mTORC2 (Rictor KO). mTORC2 suppression accelerated aging of the sperm DNA methylome and resulted in a reproductive phenotype concordant with older age, including decreased testes weight and sperm counts, and increased percent of morphologically abnormal spermatozoa and mitochondrial DNA copy number. Suppression of mTORC1 resulted in the shift of DNA methylome in sperm opposite to the shift associated with physiological aging - sperm DNA methylome rejuvenation and mild changes in sperm parameters. These results demonstrate for the first time that the balance of mTOR complexes in Sertoli cells regulates the rate of sperm epigenetic aging. Thus, mTOR pathway in Sertoli cells may be used as a novel target of therapeutic interventions to rejuvenate the sperm epigenome in advanced-age fathers.


Subject(s)
DNA Methylation , Sertoli Cells , Spermatozoa , Male , Animals , Sertoli Cells/metabolism , Mice , Spermatozoa/metabolism , Spermatozoa/physiology , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , TOR Serine-Threonine Kinases/metabolism , Mice, Knockout , Mechanistic Target of Rapamycin Complex 2/metabolism , Mechanistic Target of Rapamycin Complex 2/genetics , Regulatory-Associated Protein of mTOR/metabolism , Regulatory-Associated Protein of mTOR/genetics , Mice, Transgenic , Aging/physiology , Aging/genetics , Signal Transduction , Rapamycin-Insensitive Companion of mTOR Protein/metabolism , Rapamycin-Insensitive Companion of mTOR Protein/genetics , Epigenesis, Genetic
12.
Sci Rep ; 14(1): 21420, 2024 09 13.
Article in English | MEDLINE | ID: mdl-39271694

ABSTRACT

Both the ε4 variant of the apolipoprotein E (APOE) gene and hearing loss are well-known risk factors for Alzheimer's disease. However, previous studies have produced inconsistent findings regarding the association between APOE genotypes and hearing levels, necessitating further investigation. The aim of this study was to investigate the relationship between APOE genotypes and hearing levels. This retrospective study analyzed clinical data from a clinical data warehouse of seven affiliated Catholic Medical Center hospitals. The study included 1,162 participants with records of APOE genotypes, audiometric tests, and cognitive function tests. In Generalized linear mixed model analysis, ε4 carriers exhibited lower pure tone audiometry thresholds with an estimate of -0.353 (SE = 0.126, p = 0.005). However, the interaction term for age and APOE ε4 had a coefficient of 0.577 (SE = 0.214 p = 0.006), suggesting that the APOE ε4 gene may accelerate hearing deterioration with age. Subgroup analysis based on an age cut-off of 75 revealed that ε4 carriers had better hearing at younger ages, but showed no significant difference at older ages. These results indicate that the ε4 allele may have a biphasic effect on hearing levels depending on age.


Subject(s)
Alleles , Apolipoprotein E4 , Hearing Loss , Humans , Male , Female , Aged , Apolipoprotein E4/genetics , Retrospective Studies , Middle Aged , Hearing Loss/genetics , Aged, 80 and over , Genotype , Audiometry, Pure-Tone , Presbycusis/genetics , Aging/genetics
13.
Cells ; 13(17)2024 Aug 25.
Article in English | MEDLINE | ID: mdl-39272993

ABSTRACT

Ovarian aging results in reproductive disorders and infertility in mammals. Previous studies have reported that the ferroptosis and autophagy caused by oxidative stress may lead to ovarian aging, but the mechanisms remain unclear. In this study, we compared the morphological characteristics between the aged and young ovaries of pigs and found that the aged ovaries were larger in size and showed more corpora lutea. TUNEL assay further showed that the apoptosis level of granulosa cells (GCs) was relatively higher in the aged ovaries than those in young ovaries, as well as the expressions of autophagy-associated genes, e.g., p62, ATG7, ATG5, and BECN1, but that the expressions of oxidative stress and aging-associated genes, e.g., SOD1, SIRT1, and SIRT6, were significantly lower. Furthermore, the RNA-seq, Western blotting, and immunofluorescence suggested that phospholipid phosphatase 3 (PLPP3) protein was significantly upregulated in the aged ovaries. PLPP3 was likely to decrease the expressions of SIRT1 and SIRT6 to accelerate cellular senescence of porcine GCs, inhibit the expressions of SOD1, CAT, FSP1, FTH1, and SLC7A11 to exacerbate oxidative stress and ferroptosis, and arouse autophagy to retard the follicular development. In addition, two SNPs of PLPP3 promoter were significantly associated with the age at puberty. g.155798586 (T/T) and g.155798718 (C/C) notably facilitated the mRNA and protein level of PLPP3. In conclusion, PLPP3 might aggravate the oxidative stress of GCs to accelerate ovarian aging, and two molecular markers of PLPP3 were identified for ovarian aging in pigs. This work not only contributes to investigations on mechanisms for ovarian aging but also provides valuable molecular markers to postpone ovarian aging in populations.


Subject(s)
Aging , Granulosa Cells , Ovary , Oxidative Stress , Animals , Female , Ovary/metabolism , Ovary/pathology , Swine , Aging/genetics , Aging/metabolism , Granulosa Cells/metabolism , Autophagy/genetics , Apoptosis/genetics , Cellular Senescence/genetics , Phosphatidate Phosphatase/metabolism , Phosphatidate Phosphatase/genetics
14.
Int J Mol Sci ; 25(17)2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39273460

ABSTRACT

Degenerative diseases oftentimes occur within the continuous process of aging, and the corresponding clinical manifestations may be neurodegeneration, neoplastic diseases, or various human complex diseases. DNA methylation provides the opportunity to explore aging and degenerative diseases as epigenetic traits. It has already been applied to age prediction and disease diagnosis. It has been shown that various degenerative diseases share co-physiology mechanisms with each other, clues of which may be gained from studying the aging process. Here, we endeavor to predict the risk of degenerative diseases in an aging-relevant comorbid mechanism perspective. Firstly, an epigenetic clock method was implemented based on a multi-scale convolutional neural network, and a Shapley feature attribution analysis was applied to discover the aging-related CpG sites. Then, these sites were further screened to a smaller subset composed of 196 sites by using biomics analysis according to their biological functions and mechanisms. Finally, we constructed a multilayer perceptron (MLP)-based degenerative disease risk prediction model, Mlp-DDR, which was well trained and tested to accurately classify nine degenerative diseases. Recent studies also suggest that DNA methylation plays a significant role in conditions like osteoporosis and osteoarthritis, broadening the potential applications of our model. This approach significantly advances the ability to understand degenerative diseases and represents a substantial shift from traditional diagnostic methods. Despite the promising results, limitations regarding model complexity and dataset diversity suggest directions for future research, including the development of tissue-specific epigenetic clocks and the inclusion of a wider range of diseases.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Neurodegenerative Diseases , Humans , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/diagnosis , CpG Islands , Aging/genetics , Neural Networks, Computer
15.
Nat Commun ; 15(1): 8093, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39285199

ABSTRACT

We describe a framework that addresses concern that the rate of change in any aging biomarker displays a trivial inverse relation with maximum lifespan. We apply this framework to methylation data from the Mammalian Methylation Consortium. We study the relationship of lifespan with the average rate of change in methylation (AROCM) from two datasets: one with 90 dog breeds and the other with 125 mammalian species. After examining 54 chromatin states, we conclude three key findings: First, a reciprocal relationship exists between the AROCM in bivalent promoter regions and maximum mammalian lifespan: AROCM ∝ 1/MaxLifespan. Second, the correlation between average methylation and age bears no relation to maximum lifespan, Cor(Methyl,Age) ⊥ MaxLifespan. Third, the rate of methylation change in young animals is related to that in old animals: Young animals' AROCM ∝ Old AROCM. These findings critically hinge on the chromatin context, as different results emerge in other chromatin contexts.


Subject(s)
Chromatin , DNA Methylation , Longevity , Mammals , Promoter Regions, Genetic , Animals , Longevity/genetics , Mammals/genetics , Dogs , Chromatin/metabolism , Chromatin/genetics , Promoter Regions, Genetic/genetics , Aging/genetics , Aging/physiology , Humans
16.
Zhonghua Liu Xing Bing Xue Za Zhi ; 45(9): 1291-1301, 2024 Sep 10.
Article in Chinese | MEDLINE | ID: mdl-39307704

ABSTRACT

Aging, a process of functional decline with the increase of chronological age, is a major risk factor for chronic diseases. Aging shows significant individual differences, which is influenced by both genetic and environmental factors. Accurate measurement of physiological age helps identify individuals with accelerated aging and those at high risk for chronic diseases and mortality, which would promote individual health management and precision medicine for healthy aging. In this paper, we summarize the omics-based aging clocks and discuss their current and future applications.


Subject(s)
Aging , Humans , Aging/genetics , Biological Clocks/genetics , Genomics , Proteomics , Chronic Disease , Metabolomics
17.
Nature ; 633(8030): 608-614, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39261734

ABSTRACT

Human genetic studies of common variants have provided substantial insight into the biological mechanisms that govern ovarian ageing1. Here we report analyses of rare protein-coding variants in 106,973 women from the UK Biobank study, implicating genes with effects around five times larger than previously found for common variants (ETAA1, ZNF518A, PNPLA8, PALB2 and SAMHD1). The SAMHD1 association reinforces the link between ovarian ageing and cancer susceptibility1, with damaging germline variants being associated with extended reproductive lifespan and increased all-cause cancer risk in both men and women. Protein-truncating variants in ZNF518A are associated with shorter reproductive lifespan-that is, earlier age at menopause (by 5.61 years) and later age at menarche (by 0.56 years). Finally, using 8,089 sequenced trios from the 100,000 Genomes Project (100kGP), we observe that common genetic variants associated with earlier ovarian ageing associate with an increased rate of maternally derived de novo mutations. Although we were unable to replicate the finding in independent samples from the deCODE study, it is consistent with the expected role of DNA damage response genes in maintaining the genetic integrity of germ cells. This study provides evidence of genetic links between age of menopause and cancer risk.


Subject(s)
Aging , Genetic Predisposition to Disease , Menopause , Mutation Rate , Neoplasms , Ovary , Adult , Female , Humans , Male , Middle Aged , Aging/genetics , Aging/pathology , DNA Damage/genetics , Fertility/genetics , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Genome, Human/genetics , Germ-Line Mutation/genetics , Menarche/genetics , Menopause/genetics , Neoplasms/genetics , Ovary/metabolism , Ovary/pathology , Time Factors , UK Biobank , United Kingdom/epidemiology
18.
Nat Aging ; 4(9): 1171, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39232113
19.
Proc Natl Acad Sci U S A ; 121(37): e2416630121, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39236237
20.
PLoS One ; 19(9): e0310219, 2024.
Article in English | MEDLINE | ID: mdl-39259742

ABSTRACT

Nucleostemin (NS) plays a role in liver regeneration, and aging reduces its expression in the baseline and regenerating livers following 70% partial hepatectomy (PHx). Here we interrogate the mechanism controlling NS expression during liver regeneration and aging. The NS promoter was analyzed by TRANSFAC. Functional studies were performed using cell-based luciferase assay, endogenous NS expression in Hep3B cells, mouse livers with a gain-of-function mutation of C/EBPα (S193D), and mouse livers with C/EBPα knockdown. We found a CAAT box with four C/EBPα binding sites (-1216 to -735) and a GC box with consensus binding sites for c-Myc, E2F1, and p300-associated protein complex (-633 to -1). Age-related changes in NS expression correlated positively with the expression of c-Myc, E2F1, and p300, and negatively with that of C/EBPα and C/EBPß. PHx upregulated NS expression at 1d, coinciding with an increase in E2F1 and a decrease in C/EBPα. C/EBPα bound to the consensus sequences found in the NS promoter in vitro and in vivo, inhibited its transactivational activity in a binding site-dependent manner, and decreased the expression of endogenous NS in Hep3B cells. In vivo activation of C/EBPα by the S193D mutation resulted in a 4th-day post-PHx reduction of NS, a feature shared by 16-m/o livers. Finally, C/EBPα knockdown increased its expression in aged (24-m/o) livers under both baseline and regeneration conditions. This study reports the C/EBPα suppression of NS expression in aged livers, providing a new perspective on the mechanistic orchestration of tissue homeostasis in aging.


Subject(s)
Aging , GTP-Binding Proteins , Liver Regeneration , Nuclear Proteins , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc , Animals , Liver Regeneration/genetics , Liver Regeneration/physiology , Mice , Aging/metabolism , Aging/physiology , Aging/genetics , Humans , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , GTP-Binding Proteins/metabolism , GTP-Binding Proteins/genetics , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , CCAAT-Enhancer-Binding Protein-beta/genetics , E2F1 Transcription Factor/metabolism , E2F1 Transcription Factor/genetics , Hepatectomy , Binding Sites , Liver/metabolism , E1A-Associated p300 Protein/metabolism , Gene Expression Regulation , Transcription, Genetic , CCAAT-Enhancer-Binding Protein-alpha/metabolism , CCAAT-Enhancer-Binding Protein-alpha/genetics , Male , Carrier Proteins/metabolism , Carrier Proteins/genetics , Mice, Inbred C57BL , Cell Line, Tumor , RNA-Binding Proteins
SELECTION OF CITATIONS
SEARCH DETAIL