11
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Epigenetic alterations in longevity regulators, reduced life span, and exacerbated aging-related pathology in old father offspring mice

      research-article
      a , a , a , a , c , d , e , e , a , b , a , d , f , d , g , d , h , e ,   d , e , i , d , j , f , k , l , b , g , d , d , e , m , n , d , h , j , a , 5
      Proceedings of the National Academy of Sciences of the United States of America
      National Academy of Sciences
      aging, epigenetics, intergenerational inheritance, sperm, mTOR

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Significance

          Aging-associated diseases are increasingly common in an aging global population. However, the contributors and origins of differential risk for unhealthy aging remain poorly understood. Using a mouse model, we found that offspring of aged fathers exhibited a reduced life span and more pronounced aging-associated pathologies than animals sired by young fathers. Tissue of offspring and aged fathers revealed shared epigenetic signatures and showed altered activation states of longevity-related cell signaling. Our results suggest that variability in aging trajectories could derive, in part, from the age at conception of the father, a possibility that warrants human epidemiological investigation.

          Abstract

          Advanced age is not only a major risk factor for a range of disorders within an aging individual but may also enhance susceptibility for disease in the next generation. In humans, advanced paternal age has been associated with increased risk for a number of diseases. Experiments in rodent models have provided initial evidence that paternal age can influence behavioral traits in offspring animals, but the overall scope and extent of paternal age effects on health and disease across the life span remain underexplored. Here, we report that old father offspring mice showed a reduced life span and an exacerbated development of aging traits compared with young father offspring mice. Genome-wide epigenetic analyses of sperm from aging males and old father offspring tissue identified differentially methylated promoters, enriched for genes involved in the regulation of evolutionarily conserved longevity pathways. Gene expression analyses, biochemical experiments, and functional studies revealed evidence for an overactive mTORC1 signaling pathway in old father offspring mice. Pharmacological mTOR inhibition during the course of normal aging ameliorated many of the aging traits that were exacerbated in old father offspring mice. These findings raise the possibility that inherited alterations in longevity pathways contribute to intergenerational effects of aging in old father offspring mice.

          Related collections

          Most cited references66

          • Record: found
          • Abstract: found
          • Article: not found

          Sperm tsRNAs contribute to intergenerational inheritance of an acquired metabolic disorder.

          Increasing evidence indicates that metabolic disorders in offspring can result from the father's diet, but the mechanism remains unclear. In a paternal mouse model given a high-fat diet (HFD), we showed that a subset of sperm transfer RNA-derived small RNAs (tsRNAs), mainly from 5' transfer RNA halves and ranging in size from 30 to 34 nucleotides, exhibited changes in expression profiles and RNA modifications. Injection of sperm tsRNA fractions from HFD males into normal zygotes generated metabolic disorders in the F1 offspring and altered gene expression of metabolic pathways in early embryos and islets of F1 offspring, which was unrelated to DNA methylation at CpG-enriched regions. Hence, sperm tsRNAs represent a paternal epigenetic factor that may mediate intergenerational inheritance of diet-induced metabolic disorders.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Meta-analysis of age-related gene expression profiles identifies common signatures of aging.

            Numerous microarray studies of aging have been conducted, yet given the noisy nature of gene expression changes with age, elucidating the transcriptional features of aging and how these relate to physiological, biochemical and pathological changes remains a critical problem. We performed a meta-analysis of age-related gene expression profiles using 27 datasets from mice, rats and humans. Our results reveal several common signatures of aging, including 56 genes consistently overexpressed with age, the most significant of which was APOD, and 17 genes underexpressed with age. We characterized the biological processes associated with these signatures and found that age-related gene expression changes most notably involve an overexpression of inflammation and immune response genes and of genes associated with the lysosome. An underexpression of collagen genes and of genes associated with energy metabolism, particularly mitochondrial genes, as well as alterations in the expression of genes related to apoptosis, cell cycle and cellular senescence biomarkers, were also observed. By employing a new method that emphasizes sensitivity, our work further reveals previously unknown transcriptional changes with age in many genes, processes and functions. We suggest these molecular signatures reflect a combination of degenerative processes but also transcriptional responses to the process of aging. Overall, our results help to understand how transcriptional changes relate to the process of aging and could serve as targets for future studies. http://genomics.senescence.info/uarrays/signatures.html. Supplementary data are available at Bioinformatics online.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Linking functional decline of telomeres, mitochondria and stem cells during ageing.

              The study of human genetic disorders and mutant mouse models has provided evidence that genome maintenance mechanisms, DNA damage signalling and metabolic regulation cooperate to drive the ageing process. In particular, age-associated telomere damage, diminution of telomere 'capping' function and associated p53 activation have emerged as prime instigators of a functional decline of tissue stem cells and of mitochondrial dysfunction that adversely affect renewal and bioenergetic support in diverse tissues. Constructing a model of how telomeres, stem cells and mitochondria interact with key molecules governing genome integrity, 'stemness' and metabolism provides a framework for how diverse factors contribute to ageing and age-related disorders.
                Bookmark

                Author and article information

                Journal
                Proc Natl Acad Sci U S A
                Proc. Natl. Acad. Sci. U.S.A
                pnas
                pnas
                PNAS
                Proceedings of the National Academy of Sciences of the United States of America
                National Academy of Sciences
                0027-8424
                1091-6490
                6 March 2018
                21 February 2018
                21 February 2018
                : 115
                : 10
                : E2348-E2357
                Affiliations
                [1] aMolecular and Cellular Cognition Lab, German Center for Neurodegenerative Diseases (DZNE) , 53127 Bonn, Germany;
                [2] bSelective Vulnerability of Neurodegenerative Diseases Lab, German Center for Neurodegenerative Diseases (DZNE) , 53127 Bonn, Germany;
                [3] cInstitute of Pathology, Helmholtz Zentrum München, German Research Center for Environmental Health , 85764 Neuherberg, Germany;
                [4] dGerman Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München , 85764 Neuherberg, Germany;
                [5] eComputational Systems Biology Lab, German Center for Neurodegenerative Diseases (DZNE) , 37077 Göttingen, Germany;
                [6] fInstitute for Medical Microbiology, Immunology and Hygiene, Technische Universität München , 81675 Munich, Germany;
                [7] gChair of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-Universität München , 81377 Munich, Germany;
                [8] hGerman Center for Diabetes Research (DZD) , 85764 München-Neuherberg, Germany;
                [9] iResearch Group Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices , 53175 Bonn, Germany;
                [10] jChair of Experimental Genetics, Center of Life and Food Sciences Weihenstephan, Technische Universität München , 85350 Freising-Weihenstephan, Germany;
                [11] kDepartment of Internal Medicine I, University Hospital Carl Gustav Carus, Technical University Dresden , 01307 Dresden, Germany;
                [12] lJules Stein Eye Institute, University of California, Los Angeles , CA 90095;
                [13] mGerman Center for Neurodegenerative Diseases (DZNE) , 72076 Tübingen, Germany;
                [14] nCenter for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf , 20251 Hamburg, Germany
                Author notes
                5To whom correspondence should be addressed. Email: dan.ehninger@ 123456dzne.de .

                Edited by Nahum Sonenberg, McGill University, Montreal, QC, Canada, and approved January 24, 2018 (received for review June 2, 2017)

                Author contributions: M.W., J.A., D.H.B., G.E., W.S.J., E.W., H.F., V.G.-D., S.B., M.H.d.A., and D.E. designed research; K.X., D.P.R., B.L.P., K.S.H., F.N., R.O.V., M.H., I.L., M.S., S.S., T.A., B.R., J.R., A.-L.S., C.P., M.E.M., A.M., and D.E. performed research; K.X., D.P.R., B.L.P., K.S.H., F.N., R.O.V., M.H., M.S., T.A., B.R., J.R., C.P., M.E.M., A.M., and D.E. analyzed data; and D.P.R., B.L.P., S.B., and D.E. wrote the paper.

                1K.X., D.P.R., and B.L.P. contributed equally to this work.

                2Present address: Bioinformatics Unit, Max Planck Institute for Immunobiology and Epigenetics, 79108 Freiburg, Germany.

                3Present address: Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY 10032.

                4Present address: Institute of Stem Cell Research, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany.

                Author information
                http://orcid.org/0000-0003-1239-0547
                http://orcid.org/0000-0002-0430-9510
                http://orcid.org/0000-0003-4366-5662
                http://orcid.org/0000-0001-7464-1335
                Article
                201707337
                10.1073/pnas.1707337115
                5877957
                29467291
                404e4a46-dd8f-4a86-95a1-99d81f7b523a
                Copyright © 2018 the Author(s). Published by PNAS.

                This open access article is distributed under Creative Commons Attribution-NonCommercial-NoDerivatives License 4.0 (CC BY-NC-ND).

                History
                Page count
                Pages: 10
                Categories
                PNAS Plus
                Biological Sciences
                Medical Sciences
                PNAS Plus

                aging,epigenetics,intergenerational inheritance,sperm,mtor
                aging, epigenetics, intergenerational inheritance, sperm, mtor

                Comments

                Comment on this article