Category: Home

Caloric restriction and DNA damage

caloric restriction and DNA damage

Csloric growth damgae axis hypothesis states that increased signaling through these pathways Alternate-day fasting and hunger management the aging damagf by promoting cell growth and proliferation. Qnd caloric restriction and DNA damage for enhanced caloric restriction and DNA damage the role of novel dietary strategies in the present obesogenic environment. Mechanisms of life span extension by rapamycin in the fruit fly Drosophila melanogaster. Using DNA methylation profiling to evaluate biological age and longevity interventions. It is time to embrace 21st-century medicine. Sirtuin activators mimic caloric restriction and delay ageing in metazoans. Read More Accept.

Video

Caloric Restriction - latest human data Previous admage have shown that caloric restriction decreases mitochondrial oxygen radical production and restrcition DNA damage in rat organs, which can calotic linked to the slowing of aging rate induced by this regime. Damags two characteristics are Resupply delivery services typical snd long-lived caloric restriction and DNA damage. However, it has never been investigated if those decreases are linked to the decrease in the intake of calories themselves or to decreases in specific dietary components. In this study the possible role of the dietary protein was investigated. The decreases in ROS generation occurred specifically at complex~I. They also occurred without changes in mitochondrial oxygen consumption. Instead, there was a decrease in the percent free radical leak the percentage of total electron flow leading to ROS generation in the respiratory chain.

Caloric restriction and DNA damage -

White adipose tissue WAT forms an endocrine organ with both positive and negative effects on metabolism. By secreting adipokines , adipocytes regulate metabolism, energy intake, and fat storage. Adipocytes are known to enlarge during obesity and the ageing process. Several studies demonstrated that increased fat cell size is a significant predictor of altered blood lipid profiles and glucose-insulin homeostasis.

The contribution of visceral adiposity to these associations seems to be of particular importance. Senescence and inflammation are two important mechanisms contributing to ageing and the metabolic consequences of obesity. Inflammation can result from accumulation of macrophages in adipose tissue via production of cytokines such as TNFα and IL Increase in lipolysis has been shown to induce macrophage migration in vitro.

Macrophage numbers in adipose tissue also increase with obesity and ageing where they scavenge dead or senescent adipocytes.

However, inflammatory cytokines and chemokines are also characteristics of the senescence-associated secretory phenotype SASP in senescent cells.

We have shown previously that reactive oxygen species ROS , DNA damage , and mitochondrial dysfunction are instrumental to maintain cellular senescence. Various treatments have been suggested to delay senescence in adipose tissues while obesity and short telomeres exacerbated senescence.

A recent study showed that feeding a high-fat diet ad libitum induced senescence in mouse visceral adipose tissue which could be ameliorated by exercise. However, dietary restriction DR seems to regulate many more genes than exercise in subcutaneous fat in humans.

We have demonstrated previously that short-term dietary restriction in wild type mice decreased the amount of senescent cells in various tissues. We hypothesise that pro-inflammatory cytokines and senescence are also causally related in visceral WAT, increase together during ageing, and might be rescued during DR.

We used visceral WAT from mice of different ages as well as mice on late-onset, short term DR to investigate the changes in adipocyte size, accumulation of DNA damage during ageing and DR, together with the expression of pro-inflammatory cytokines TNFα, IL-6, IL-1β , and senescence markers p16 and p We also analysed AMPK activity which is an important signal transduction pathway implicated in the regulation of physiological processes of DR.

AMPK activation is thought to be able to inhibit inflammatory responses and plays a central role in the regulation of whole body energy homeostasis and functions as a key regulator of intracellular fatty acid metabolism. Our results demonstrate increased senescence and inflammation during ageing in mouse visceral fat while DR was able to ameliorate several of these parameters.

DR was able to significantly reduce adipocyte size and multiple markers of adipocyte senescence significant for DNA damage, p21 and IL-6 expression.

This indicates that DR acts as a senolytic treatment in visceral fat, similar to its effects in other tissues. This highlights the health benefits of a decreased nutritional intake over a relatively short period of time at middle age. Now we need a correct protocol of DR,CR or outright fasting.

Intuitively, it seems that CR can stimulate the autophagy, which can prevent pre-senescsant cells beginning senescent and even killing some weaker cells who don't have get enough energy. On the other hand the human body is easy to complex and the there might be some negative effects too.

That's why a correct portfolio is so needed. I've fasted four times using the Prolon diet over the last 15 months. I don't have any quotes, but I have read somewhere where Valter Longo states that fasting-mimicking diets of five days do clear out stubborn body fat that accumulates with age.

Maybe the stomach fat cells perform apoptosis? Genetically, it would be more beneficial for you if you are homozygous for the good alleles of TNF-a, IL-6, IL-1b, and AMPK genes. I checked my genome and am homozygous for the beneficial SNP allele of all 4 of these genes. About 10 ago I did a 22 day fast and lost closer to 50 pounds.

IV regression is a method commonly used to reduce the impact of confounding in association analysis. Under conditions of nonadherence, traditional ITT analysis can result in a biased estimate of the treatment effect and an IV estimator can provide a complement The ITT estimate may therefore underestimate the effect of CR on biological aging.

The IV approach we used involved two related regressions. The second regression modeled the outcomes changes in measures of biological aging as functions of the predicted treatment dose estimated by the first regression and pretreatment covariates.

The base first-stage regression took the form. Results from this first-stage regression were then included in the second-stage model:.

For final TOT analysis, we included a further instrument in the first-stage regression consisting of the interaction between the baseline level of the aging measure and the CR treatment group. Sensitivity analysis involving re-estimating the IV regression models omitting this final instrument did not change results.

Supplementary Fig. Data met model assumptions. Normality of outcome variables was evaluated by visual inspection of distributions and the Shapiro—Wilk test Equality of variances was evaluated according to the tests proposed by Brown and Forsythe 68 and Markowski and Markowski Models used to test ITT and TOT effects were fitted with heteroskedasticity-robust standard errors.

Normality of distribution of error terms was evaluated by visual inspection of histograms of residuals and the Shapiro—Wilk test.

The clocks we analyzed were developed to predict mortality risk. The age values computed by the clock algorithms correspond to the age at which predicted mortality risk would be approximately normal in the reference population used to develop the clock.

Pace-of-aging measures estimate the rate of biological aging, defined as the rate of decline in overall system integrity. Pace-of-aging values correspond to the years of biological aging experienced during a single calendar year. A value of 1 represents the typical pace of aging in a reference population; values above 1 indicate faster pace of aging; values below 1 indicate slower pace of aging.

Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article. Applications for some types of data may require IRB oversight. Source data for Fig. Kaeberlein, M. Longevity and aging. FPrime Rep.

Kennedy, B. et al. Geroscience: linking aging to chronic disease. Cell , — Article CAS PubMed PubMed Central Google Scholar. Campisi, J. From discoveries in ageing research to therapeutics for healthy ageing.

Nature , — Speakman, J. Caloric restriction. Aspects Med. Article CAS PubMed Google Scholar. Maegawa, S. Caloric restriction delays age-related methylation drift. Article PubMed PubMed Central Google Scholar. Hahn, O. Dietary restriction protects from age-associated DNA methylation and induces epigenetic reprogramming of lipid metabolism.

Genome Biol. Petkovich, D. Using DNA methylation profiling to evaluate biological age and longevity interventions. Cell Metab. e6 Anderson, R. The caloric restriction paradigm: implications for healthy human aging.

Mattison, J. Impact of caloric restriction on health and survival in rhesus monkeys from the NIA study. Ravussin, E. A 2-year randomized controlled trial of human caloric restriction: feasibility and effects on predictors of health span and longevity. A 70 , — Article CAS Google Scholar.

Scott, A. The economic value of targeting aging. Aging 1 , — It is time to embrace 21st-century medicine. Public Policy Aging Rep. Article Google Scholar. Goldman, D. Substantial health and economic returns from delayed aging may warrant a new focus for medical research.

Health Aff. Fahy, G. Reversal of epigenetic aging and immunosenescent trends in humans. Aging Cell 18 , e Chen, L. Effects of vitamin D3 supplementation on epigenetic aging in overweight and obese African Americans with suboptimal vitamin D status: a randomized clinical trial.

A 74 , 91—98 Sae-Lee, C. Dietary intervention modifies DNA methylation age assessed by the epigenetic clock. Food Res. Colchero, F.

Fleming, T. Biomarkers and surrogate endpoints in clinical trials. Prentice, R. Surrogate endpoints in clinical trials: definition and operational criteria.

Justice, J. A framework for selection of blood-based biomarkers for geroscience-guided clinical trials: report from the TAME Biomarkers Workgroup. GeroScience 40 , — Racette, S. Levine, M. An epigenetic biomarker of aging for lifespan and healthspan. Aging 10 , — Lu, A.

DNA methylation GrimAge strongly predicts lifespan and healthspan. Aging 11 , — Belsky, D. DunedinPACE, a DNA methylation biomarker of the pace of aging. eLife 11 , e Higgins-Chen, A. A computational solution for bolstering reliability of epigenetic clocks: implications for clinical trials and longitudinal tracking.

Aging 2 , — Kraus, W. Lancet Diabetes Endocrinol. Benjamin, D. Redefine statistical significance. Article PubMed Google Scholar. Salas, L. An optimized library for reference-based deconvolution of whole-blood biospecimens assayed using the Illumina HumanMethylationEPIC BeadArray.

Sugden, K. Association of pace of aging measured by blood-based DNA methylation with age-related cognitive impairment and dementia.

Neurology 99 , e—e Hillary, R. Epigenetic measures of ageing predict the prevalence and incidence of leading causes of death and disease burden. Epigenetics 12 , Change in the rate of biological aging in response to caloric restriction: CALERIE Biobank Analysis. A 73 , 4—10 Kwon, D. A toolkit for quantification of biological age from blood chemistry and organ function test data: BioAge.

Geroscience 43 , — López-Otín, C. The hallmarks of aging. Spadaro, O. Caloric restriction in humans reveals immunometabolic regulators of health span. Science , — Redman, L. Metabolic slowing and reduced oxidative damage with sustained caloric restriction support the rate of living and oxidative damage theories of aging.

e4 Anthonisen, N. The effects of a smoking cessation intervention on Ferrucci, L. Measuring biological aging in humans: a quest.

Aging Cell 19 , e Kritchevsky, S. Testing the geroscience hypothesis: early days. A 75 , 99— Bell, C. DNA methylation aging clocks: challenges and recommendations. Eleven telomere, epigenetic clock, and biomarker-composite quantifications of biological aging: do they measure the same thing?

PubMed Google Scholar. Ahadi, S. Personal aging markers and ageotypes revealed by deep longitudinal profiling. Ma, S. Caloric restriction reprograms the single-cell transcriptional landscape of Rattus norvegicus aging.

Cell , — e22 Dorling, J. Calorie restriction for enhanced longevity: the role of novel dietary strategies in the present obesogenic environment. Ageing Res.

Das, S. Body-composition changes in the Comprehensive Assessment of Long-term Effects of Reducing Intake of Energy CALERIE -2 study: a 2-y randomized controlled trial of calorie restriction in nonobese humans.

Shen, W. Effect of 2-year caloric restriction on organ and tissue size in nonobese to year-old adults in a randomized clinical trial: the CALERIE study. Moffitt, T.

The longitudinal study of aging in human young adults: knowledge gaps and research agenda. A 72 , — Sierra, F. Moving geroscience from the bench to clinical care and health policy. Frameworks for proof-of-concept clinical trials of interventions that target fundamental aging processes.

A Biol. Putting epigenetic biomarkers to the test for clinical trials. eLife 9 , e Longo, V. Intermittent and periodic fasting, longevity and disease. Aging 1 , 47—59 Davis, S. methylumi: handle Illumina methylation data. R package version 2. Huber, W. Orchestrating high-throughput genomic analysis with Bioconductor.

Methods 12 , — Lehne, B. A coherent approach for analysis of the Illumina HumanMethylation BeadChip improves data quality and performance in epigenome-wide association studies.

Aryee, M. Minfi: a flexible and comprehensive Bioconductor package for the analysis of Infinium DNA methylation microarrays.

Bioinformatics 30 , — Horvath, S. DNA methylation-based biomarkers and the epigenetic clock theory of ageing. DNA methylation age of human tissues and cell types. Hannum, G. Genome-wide methylation profiles reveal quantitative views of human aging rates.

Cell 49 , — Chen, B. DNA methylation-based measures of biological age: meta-analysis predicting time to death. Aging 8 , — Assessment of epigenetic clocks as biomarkers of aging in basic and population research. A 75 , — Quantification of biological aging in young adults.

Natl Acad. USA , E—E Elliott, M. Disparities in the pace of biological aging among midlife adults of the same chronological age have implications for future frailty risk and policy.

Patterns of reliability: assessing the reproducibility and integrity of DNA methylation measurement. Patterns 1 , Stata Multilevel Mixed-Effects Reference Manual StataCorp, Sussman, J. An IV for the RCT: using instrumental variables to adjust for treatment contamination in randomised controlled trials.

Baum, C. Instrumental variables and GMM: estimation and testing. Stata J. Bang, H. On estimating treatment effects under non-compliance in randomized clinical trials: are intent-to-treat or instrumental variables analyses perfect solutions?

Shapiro, S. An analysis of variance test for normality complete samples. Biometrika 52 , — Brown, M. Robust tests for the equality of variances. Markowski, C. Conditions for the effectiveness of a preliminary test of variance. Google Scholar. Download references. This research was supported by grant no.

R01AG to D. received additional support from the American Brain Foundation to R. and V. received additional support from grant no. P30AG to C. R01AG to V. and C. R33AG to K. received additional support from the CIHR grant no. RN to M.

and S. received support from grant no. R01 AG to S. and W. R03AG to I. U01AG to B. We thank the CALERIE Research Network no. R33AG for their assistance in this project and the Dunedin Study no. R01AG for facilitating early access to the DunedinPACE DNA methylation algorithm.

The funders had no role in study design, data collection and analysis, decision to publish or preparation of the paper. completed work on this project while affiliated with the Butler Columbia Aging Center.

She is now in the Department of Neurology at the Columbia University Irving Medical Center. Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY, USA.

Waziry, C. Ryan, M. Kothari, G. Department of Genetics, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA. Duke Molecular Physiology Institute and Department of Medicine, Duke University School of Medicine, Durham, NC, USA. Huffman, V.

Department of Medical Genetics, Edwin S. Leong Healthy Aging Program, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada.

Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY, USA. Center on Aging and Development, Biostatistics and Bioinformatics, Duke University, Durham, NC, USA.

Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA. Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA. Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.

Department of Biobehavioral Health, Pennsylvania State University, State College, PA, USA. Pennington Biomedical Research Center, Baton Rouge, LA, USA.

Department of Medicine, Duke University School of Medicine, Durham, NC, USA. Program in Physical Therapy and Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA. College of Health Solutions, Arizona State University, Phoenix, AZ, USA.

Buck Institute for Research on Aging, Novato, CA, USA. You can also search for this author in PubMed Google Scholar. designed the research. Kebbe, D. and B. conducted the research. and D. prepared the DNA methylation datasets.

analyzed the data. and R. wrote the first draft of the paper. wrote the revised draft of the paper. All authors contributed critical review of the paper. Correspondence to D. are listed as inventors on a Duke University and University of Otago invention, DunedinPACE, that was licensed to a commercial entity.

The other authors declare no competing interests. Nature Aging thanks the anonymous reviewers for their contribution to the peer review of this work. Effects estimates of CR treatment from mixed models of change in epigenetic age used in Supplementary Fig. Effects estimates of CR treatment from mixed models of change in epigenetic age used in Fig.

Open Access This article is licensed under a Creative Commons Attribution 4. Reprints and permissions. Waziry, R. Effect of long-term caloric restriction on DNA methylation measures of biological aging in healthy adults from the CALERIE trial. Nat Aging 3 , — Download citation.

Received : 08 September Accepted : 22 December Published : 09 February Issue Date : March Anyone you share the following link with will be able to read this content:. Sorry, a shareable link is not currently available for this article. Provided by the Springer Nature SharedIt content-sharing initiative.

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily. Skip to main content Thank you for visiting nature. nature nature aging letters article. Download PDF. Subjects Predictive markers. This article has been updated.

Do dakage want to live a longer life in good health? Simple practices can make some difference, such calofic exercise or calorie Metabolism and metabolism syndrome. But over the long haul all that caloric restriction and DNA damage restrictjon is retriction caloric restriction and DNA damage medicine: building new classes of therapy to repair and reverse the known root causes of aging. The sooner these treatments arrive, the more lives will be saved. Find out how to help ». We know that calorie restriction slows the accumulation of nuclear DNA damage - possibly by enhancing DNA repair mechanisms - just as it slows more or less every other age-related change of interest that scientists have investigated. The degree to which ongoing random damage to nuclear DNA contributes to degenerative aging is debatedhowever:. caloric restriction and DNA damage

Author: Dokora

1 thoughts on “Caloric restriction and DNA damage

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com