1
|
de Magalhães JP, Abidi Z, dos Santos GA, Avelar RA, Barardo D, Chatsirisupachai K, Clark P, De-Souza EA, Johnson EJ, Lopes I, Novoa G, Senez L, Talay A, Thornton D, To P. Human Ageing Genomic Resources: updates on key databases in ageing research. Nucleic Acids Res 2024; 52:D900-D908. [PMID: 37933854 PMCID: PMC10767973 DOI: 10.1093/nar/gkad927] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/06/2023] [Accepted: 10/09/2023] [Indexed: 11/08/2023] Open
Abstract
Ageing is a complex and multifactorial process. For two decades, the Human Ageing Genomic Resources (HAGR) have aided researchers in the study of various aspects of ageing and its manipulation. Here, we present the key features and recent enhancements of these resources, focusing on its six main databases. One database, GenAge, focuses on genes related to ageing, featuring 307 genes linked to human ageing and 2205 genes associated with longevity and ageing in model organisms. AnAge focuses on ageing, longevity, and life-history across animal species, containing data on 4645 species. DrugAge includes information about 1097 longevity drugs and compounds in model organisms such as mice, rats, flies, worms and yeast. GenDR provides a list of 214 genes associated with the life-extending benefits of dietary restriction in model organisms. CellAge contains a catalogue of 866 genes associated with cellular senescence. The LongevityMap serves as a repository for genetic variants associated with human longevity, encompassing 3144 variants pertaining to 884 genes. Additionally, HAGR provides various tools as well as gene expression signatures of ageing, dietary restriction, and replicative senescence based on meta-analyses. Our databases are integrated, regularly updated, and manually curated by experts. HAGR is freely available online (https://genomics.senescence.info/).
Collapse
Affiliation(s)
- João Pedro de Magalhães
- Genomics of Ageing and Rejuvenation Lab, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2WB, UK
| | - Zoya Abidi
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| | - Gabriel Arantes dos Santos
- Genomics of Ageing and Rejuvenation Lab, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2WB, UK
| | - Roberto A Avelar
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| | - Diogo Barardo
- NOVOS Labs, 100 Park Avenue, 16th Fl, New York, NY 10017, USA
| | - Kasit Chatsirisupachai
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| | - Peter Clark
- Genomics of Ageing and Rejuvenation Lab, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2WB, UK
| | - Evandro A De-Souza
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas 13083-970, SP, Brazil
| | - Emily J Johnson
- Computational Biology Facility, Liverpool Shared Research Facilities, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7ZB, UK
| | - Inês Lopes
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| | - Guy Novoa
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| | - Ludovic Senez
- Genomics of Ageing and Rejuvenation Lab, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2WB, UK
| | - Angelo Talay
- Genomics of Ageing and Rejuvenation Lab, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2WB, UK
| | - Daniel Thornton
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| | - Paul Ka Po To
- Genomics of Ageing and Rejuvenation Lab, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2WB, UK
| |
Collapse
|
2
|
Das JK, Banskota N, Candia J, Griswold ME, Orenduff M, de Cabo R, Corcoran DL, Das SK, De S, Huffman KM, Kraus VB, Kraus WE, Martin C, Racette SB, Redman LM, Schilling B, Belsky D, Ferrucci L. Calorie restriction modulates the transcription of genes related to stress response and longevity in human muscle: The CALERIE study. Aging Cell 2023; 22:e13963. [PMID: 37823711 PMCID: PMC10726900 DOI: 10.1111/acel.13963] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/28/2023] [Accepted: 08/01/2023] [Indexed: 10/13/2023] Open
Abstract
The lifespan extension induced by 40% caloric restriction (CR) in rodents is accompanied by postponement of disease, preservation of function, and increased stress resistance. Whether CR elicits the same physiological and molecular responses in humans remains mostly unexplored. In the CALERIE study, 12% CR for 2 years in healthy humans induced minor losses of muscle mass (leg lean mass) without changes of muscle strength, but mechanisms for muscle quality preservation remained unclear. We performed high-depth RNA-Seq (387-618 million paired reads) on human vastus lateralis muscle biopsies collected from the CALERIE participants at baseline, 12- and 24-month follow-up from the 90 CALERIE participants randomized to CR and "ad libitum" control. Using linear mixed effect model, we identified protein-coding genes and splicing variants whose expression was significantly changed in the CR group compared to controls, including genes related to proteostasis, circadian rhythm regulation, DNA repair, mitochondrial biogenesis, mRNA processing/splicing, FOXO3 metabolism, apoptosis, and inflammation. Changes in some of these biological pathways mediated part of the positive effect of CR on muscle quality. Differentially expressed splicing variants were associated with change in pathways shown to be affected by CR in model organisms. Two years of sustained CR in humans positively affected skeletal muscle quality, and impacted gene expression and splicing profiles of biological pathways affected by CR in model organisms, suggesting that attainable levels of CR in a lifestyle intervention can benefit muscle health in humans.
Collapse
Affiliation(s)
- Jayanta Kumar Das
- Longitudinal Studies Section, Translation Gerontology BranchNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| | - Nirad Banskota
- Computational Biology and Genomics CoreNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| | - Julián Candia
- Longitudinal Studies Section, Translation Gerontology BranchNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| | | | - Melissa Orenduff
- Duke Molecular Physiology Institute and Department of MedicineDuke University School of MedicineDurhamNorth CarolinaUSA
| | - Rafael de Cabo
- Translation Gerontology Branch, National Institute on AgingNational Institutes of HealthBaltimoreMarylandUSA
| | - David L. Corcoran
- Department of GeneticsUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Sai Krupa Das
- Energy Metabolism, Jean Mayer USDA Human Nutrition Research Center on AgingTufts UniversityBostonMassachusettsUSA
| | - Supriyo De
- Computational Biology and Genomics CoreNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| | - Kim Marie Huffman
- Duke Molecular Physiology Institute and Department of MedicineDuke University School of MedicineDurhamNorth CarolinaUSA
| | - Virginia B. Kraus
- Duke Molecular Physiology Institute and Department of MedicineDuke University School of MedicineDurhamNorth CarolinaUSA
| | - William E. Kraus
- Duke Molecular Physiology Institute and Department of MedicineDuke University School of MedicineDurhamNorth CarolinaUSA
| | - Corby K. Martin
- Pennington Biomedical Research CenterLouisiana State UniversityBaton RougeLouisianaUSA
| | - Susan B. Racette
- College of Health SolutionsArizona State UniversityPhoenixArizonaUSA
| | - Leanne M. Redman
- Pennington Biomedical Research CenterLouisiana State UniversityBaton RougeLouisianaUSA
| | | | - Daniel W. Belsky
- Department of Epidemiology & Butler Columbia Aging CenterColumbia University Mailman School of Public HealthNew York CityNew YorkUSA
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translation Gerontology BranchNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| |
Collapse
|
3
|
van’t Sant LJ, Birkisdóttir MB, Ozinga RA, Gyenis Á, Hoeijmakers JH, Vermeij WP, Jaarsma D. Gene expression changes in cerebellum induced by dietary restriction. Front Mol Neurosci 2023; 16:1185665. [PMID: 37293544 PMCID: PMC10244750 DOI: 10.3389/fnmol.2023.1185665] [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: 03/13/2023] [Accepted: 05/03/2023] [Indexed: 06/10/2023] Open
Abstract
Background Dietary restriction (DR) is a well-established universal anti-aging intervention, and is neuroprotective in multiple models of nervous system disease, including models with cerebellar pathology. The beneficial effects of DR are associated with a rearrangement of gene expression that modulate metabolic and cytoprotective pathways. However, the effect of DR on the cerebellar transcriptome remained to be fully defined. Results Here we analyzed the effect of a classical 30% DR protocol on the transcriptome of cerebellar cortex of young-adult male mice using RNAseq. We found that about 5% of expressed genes were differentially expressed in DR cerebellum, the far majority of whom showing subtle expression changes. A large proportion of down-regulated genes are implicated in signaling pathways, in particular pathways associated with neuronal signaling. DR up regulated pathways in large part were associated with cytoprotection and DNA repair. Analysis of the expression of cell-specific gene sets, indicated a strong enrichment of DR down genes in Purkinje cells, while genes specifically associated with granule cells did not show such a preferential down-regulation. Conclusion Our data show that DR may have a clear effect on the cerebellar transcriptome inducing a mild shift from physiology towards maintenance and repair, and having cell-type specific effects.
Collapse
Affiliation(s)
| | - María B. Birkisdóttir
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Rutger A. Ozinga
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Ákos Gyenis
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine, Institute for Genome Stability in Ageing and Disease, University of Cologne, Cologne, Germany
| | - Jan H.J. Hoeijmakers
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Medicine, Institute for Genome Stability in Ageing and Disease, University of Cologne, Cologne, Germany
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Wilbert P. Vermeij
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Dick Jaarsma
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| |
Collapse
|
4
|
Ghaddar A, Mony VK, Mishra S, Berhanu S, Johnson JC, Enriquez-Hesles E, Harrison E, Patel A, Horak MK, Smith JS, O'Rourke EJ. Increased alcohol dehydrogenase 1 activity promotes longevity. Curr Biol 2023; 33:1036-1046.e6. [PMID: 36805847 PMCID: PMC10236445 DOI: 10.1016/j.cub.2023.01.059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/28/2022] [Accepted: 01/26/2023] [Indexed: 02/19/2023]
Abstract
Several molecules can extend healthspan and lifespan across organisms. However, most are upstream signaling hubs or transcription factors orchestrating complex anti-aging programs. Therefore, these molecules point to but do not reveal the fundamental mechanisms driving longevity. Instead, downstream effectors that are necessary and sufficient to promote longevity across conditions or organisms may reveal the fundamental anti-aging drivers. Toward this goal, we searched for effectors acting downstream of the transcription factor EB (TFEB), known as HLH-30 in C. elegans, because TFEB/HLH-30 is necessary across anti-aging interventions and its overexpression is sufficient to extend C. elegans lifespan and reduce biomarkers of aging in mammals including humans. As a result, we present an alcohol-dehydrogenase-mediated anti-aging response (AMAR) that is essential for C. elegans longevity driven by HLH-30 overexpression, caloric restriction, mTOR inhibition, and insulin-signaling deficiency. The sole overexpression of ADH-1 is sufficient to activate AMAR, which extends healthspan and lifespan by reducing the levels of glycerol-an age-associated and aging-promoting alcohol. Adh1 overexpression is also sufficient to promote longevity in yeast, and adh-1 orthologs are induced in calorically restricted mice and humans, hinting at ADH-1 acting as an anti-aging effector across phyla.
Collapse
Affiliation(s)
- Abbas Ghaddar
- Department of Biology, College of Arts and Sciences, University of Virginia, Charlottesville, VA 22903, USA
| | - Vinod K Mony
- Department of Biology, College of Arts and Sciences, University of Virginia, Charlottesville, VA 22903, USA
| | - Swarup Mishra
- Department of Biology, College of Arts and Sciences, University of Virginia, Charlottesville, VA 22903, USA; Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Samuel Berhanu
- Department of Biology, College of Arts and Sciences, University of Virginia, Charlottesville, VA 22903, USA
| | - James C Johnson
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Elisa Enriquez-Hesles
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Emma Harrison
- Department of Biology, College of Arts and Sciences, University of Virginia, Charlottesville, VA 22903, USA
| | - Aaroh Patel
- Department of Biology, College of Arts and Sciences, University of Virginia, Charlottesville, VA 22903, USA
| | - Mary Kate Horak
- Department of Biology, College of Arts and Sciences, University of Virginia, Charlottesville, VA 22903, USA; Department of Cell Biology, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| | - Jeffrey S Smith
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Eyleen J O'Rourke
- Department of Biology, College of Arts and Sciences, University of Virginia, Charlottesville, VA 22903, USA; Department of Cell Biology, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA; Robert M. Berne Cardiovascular Research Center, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA.
| |
Collapse
|
5
|
Shaposhnikov MV, Guvatova ZG, Zemskaya NV, Koval LA, Schegoleva EV, Gorbunova AA, Golubev DA, Pakshina NR, Ulyasheva NS, Solovev IA, Bobrovskikh MA, Gruntenko NE, Menshanov PN, Krasnov GS, Kudryavseva AV, Moskalev AA. Molecular mechanisms of exceptional lifespan increase of Drosophila melanogaster with different genotypes after combinations of pro-longevity interventions. Commun Biol 2022; 5:566. [PMID: 35681084 PMCID: PMC9184560 DOI: 10.1038/s42003-022-03524-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 05/24/2022] [Indexed: 12/13/2022] Open
Abstract
Aging is one of the global challenges of our time. The search for new anti-aging interventions is also an issue of great actuality. We report on the success of Drosophila melanogaster lifespan extension under the combined influence of dietary restriction, co-administration of berberine, fucoxanthin, and rapamycin, photodeprivation, and low-temperature conditions up to 185 days in w1118 strain and up to 213 days in long-lived E(z)/w mutants. The trade-off was found between longevity and locomotion. The transcriptome analysis showed an impact of epigenetic alterations, lipid metabolism, cellular respiration, nutrient sensing, immune response, and autophagy in the registered effect. The lifespan of fruit flies can be extended up to 213 days under specialized conditions.
Collapse
|
6
|
Fischer F, Grigolon G, Benner C, Ristow M. Evolutionarily conserved transcription factors as regulators of longevity and targets for geroprotection. Physiol Rev 2022; 102:1449-1494. [PMID: 35343830 DOI: 10.1152/physrev.00017.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Aging is the single largest risk factor for many debilitating conditions, including heart diseases, stroke, cancer, diabetes, and neurodegenerative disorders. While far from understood in its full complexity, it is scientifically well-established that aging is influenced by genetic and environmental factors, and can be modulated by various interventions. One of aging's early hallmarks are aberrations in transcriptional networks, controlling for example metabolic homeostasis or the response to stress. Evidence in different model organisms abounds that a number of evolutionarily conserved transcription factors, which control such networks, can affect lifespan and healthspan across species. These transcription factors thus potentially represent conserved regulators of longevity and are emerging as important targets in the challenging quest to develop treatments to mitigate age-related diseases, and possibly even to slow aging itself. This review provides an overview of evolutionarily conserved transcription factors that impact longevity or age-related diseases in at least one multicellular model organism (nematodes, flies, or mice), and/or are tentatively linked to human aging. Discussed is the general evidence for transcriptional regulation of aging and disease, followed by a more detailed look at selected transcription factor families, the common metabolic pathways involved, and the targeting of transcription factors as a strategy for geroprotective interventions.
Collapse
Affiliation(s)
- Fabian Fischer
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, Switzerland
| | - Giovanna Grigolon
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, Switzerland
| | - Christoph Benner
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, Switzerland
| | - Michael Ristow
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, Switzerland
| |
Collapse
|
7
|
Pandey M, Bansal S, Chawla G. Evaluation of lifespan promoting effects of biofortified wheat in Drosophila melanogaster. Exp Gerontol 2022; 160:111697. [PMID: 35016996 PMCID: PMC7613042 DOI: 10.1016/j.exger.2022.111697] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 12/15/2021] [Accepted: 01/05/2022] [Indexed: 11/04/2022]
Abstract
Evaluation of nutritionally enhanced biofortified dietary interventions that increase lifespan may uncover cost-effective and sustainable approaches for treatment of age-related morbidities and increasing healthy life expectancy. In this study, we report that anthocyanin rich, high yielding crossbred blue wheat prolongs lifespan of Drosophila melanogaster in different dietary contexts. In addition to functioning as an antioxidant rich intervention, the biofortified blue wheat also works through modulating expression of DR pathway genes including AMPK alpha, SREBP, PEPCK and Cry. Supplementation with blue- or purple-colored wheat provided better protection against paraquat-induced oxidative stress than control diet and increased survivability of flies in which superoxide dismutase 2 was knocked down conditionally in adults. Lastly, our findings indicate that supplementing biofortified blue wheat formulated diet prevented the decrease in lifespan and cardiac structural pathologies associated with intake of high fat diet. Overall, our findings indicate that plant-based diets formulated with biofortified cereal crops promote healthy ageing and delay progression of diseases that are exacerbated by accumulation of oxidative damage.
Collapse
Affiliation(s)
- Manish Pandey
- RNA Biology Laboratory, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad 121001, Haryana, India
| | - Sakshi Bansal
- RNA Biology Laboratory, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad 121001, Haryana, India
| | - Geetanjali Chawla
- RNA Biology Laboratory, Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad 121001, Haryana, India.
| |
Collapse
|
8
|
Vega Magdaleno GD, Bespalov V, Zheng Y, Freitas AA, de Magalhaes JP. Machine learning-based predictions of dietary restriction associations across ageing-related genes. BMC Bioinformatics 2022; 23:10. [PMID: 34983372 PMCID: PMC8729156 DOI: 10.1186/s12859-021-04523-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 12/08/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Dietary restriction (DR) is the most studied pro-longevity intervention; however, a complete understanding of its underlying mechanisms remains elusive, and new research directions may emerge from the identification of novel DR-related genes and DR-related genetic features. RESULTS This work used a Machine Learning (ML) approach to classify ageing-related genes as DR-related or NotDR-related using 9 different types of predictive features: PathDIP pathways, two types of features based on KEGG pathways, two types of Protein-Protein Interactions (PPI) features, Gene Ontology (GO) terms, Genotype Tissue Expression (GTEx) expression features, GeneFriends co-expression features and protein sequence descriptors. Our findings suggested that features biased towards curated knowledge (i.e. GO terms and biological pathways), had the greatest predictive power, while unbiased features (mainly gene expression and co-expression data) have the least predictive power. Moreover, a combination of all the feature types diminished the predictive power compared to predictions based on curated knowledge. Feature importance analysis on the two most predictive classifiers mostly corroborated existing knowledge and supported recent findings linking DR to the Nuclear Factor Erythroid 2-Related Factor 2 (NRF2) signalling pathway and G protein-coupled receptors (GPCR). We then used the two strongest combinations of feature type and ML algorithm to predict DR-relatedness among ageing-related genes currently lacking DR-related annotations in the data, resulting in a set of promising candidate DR-related genes (GOT2, GOT1, TSC1, CTH, GCLM, IRS2 and SESN2) whose predicted DR-relatedness remain to be validated in future wet-lab experiments. CONCLUSIONS This work demonstrated the strong potential of ML-based techniques to identify DR-associated features as our findings are consistent with literature and recent discoveries. Although the inference of new DR-related mechanistic findings based solely on GO terms and biological pathways was limited due to their knowledge-driven nature, the predictive power of these two features types remained useful as it allowed inferring new promising candidate DR-related genes.
Collapse
Affiliation(s)
- Gustavo Daniel Vega Magdaleno
- Integrative Genomics of Ageing Group, Institute of Life Course and Medical Sciences, University of Liverpool, 6 West Derby St, Liverpool, L7 8TX, UK
| | - Vladislav Bespalov
- School of Computer Technologies and Controls, ITMO University, Kronverkskiy Prospekt 49, 197101, St Petersburg, Russia
| | - Yalin Zheng
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, 6 West Derby St, Liverpool, L7 8TX, UK
| | - Alex A Freitas
- School of Computing, University of Kent, Canterbury, CT2 7NF, UK
| | - Joao Pedro de Magalhaes
- Integrative Genomics of Ageing Group, Institute of Life Course and Medical Sciences, University of Liverpool, 6 West Derby St, Liverpool, L7 8TX, UK.
| |
Collapse
|
9
|
Saito K, Ito M, Chiba T, Jia H, Kato H. A Comparison of Gene Expression Profiles of Rat Tissues after Mild and Short-Term Calorie Restrictions. Nutrients 2021; 13:2277. [PMID: 34209243 PMCID: PMC8308279 DOI: 10.3390/nu13072277] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/22/2021] [Accepted: 06/24/2021] [Indexed: 12/14/2022] Open
Abstract
Many studies have shown the beneficial effects of calorie restriction (CR) on rodents' aging; however, the molecular mechanism explaining these beneficial effects is still not fully understood. Previously, we conducted transcriptomic analysis on rat liver with short-term and mild-to-moderate CR to elucidate its early response to such diet. Here, we expanded transcriptome analysis to muscle, adipose tissue, intestine, and brain and compared the gene expression profiles of these multiple organs and of our previous dataset. Several altered gene expressions were found, some of which known to be related to CR. Notably, the commonly regulated genes by CR include nicotinamide phosphoribosyltransferase and heat shock protein 90, which are involved in declining the aging process and thus potential therapeutic targets for aging-related diseases. The data obtained here provide information on early response markers and key mediators of the CR-induced delay in aging as well as on age-associated pathological changes in mammals.
Collapse
Affiliation(s)
- Kenji Saito
- Health Nutrition, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; (K.S.); (M.I.); (H.J.)
| | - Maiko Ito
- Health Nutrition, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; (K.S.); (M.I.); (H.J.)
- Biomedical Gerontology Laboratory, Faculty of Human Sciences, Waseda University, 2-579-1 Mikajima, Tokorozawa, Saitama 359-1164, Japan;
| | - Takuya Chiba
- Biomedical Gerontology Laboratory, Faculty of Human Sciences, Waseda University, 2-579-1 Mikajima, Tokorozawa, Saitama 359-1164, Japan;
| | - Huijuan Jia
- Health Nutrition, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; (K.S.); (M.I.); (H.J.)
| | - Hisanori Kato
- Health Nutrition, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; (K.S.); (M.I.); (H.J.)
| |
Collapse
|
10
|
Kendig MD, Leigh SJ, Morris MJ. Unravelling the impacts of western-style diets on brain, gut microbiota and cognition. Neurosci Biobehav Rev 2021; 128:233-243. [PMID: 34153343 DOI: 10.1016/j.neubiorev.2021.05.031] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 03/23/2021] [Accepted: 05/24/2021] [Indexed: 02/06/2023]
Abstract
The steady rise in the prevalence of obesity has been fostered by modern environments that reduce energy expenditure and encourage consumption of 'western'-style diets high in fat and sugar. Obesity has been consistently associated with impairments in executive function and episodic memory, while emerging evidence indicates that high-fat, high-sugar diets can impair aspects of cognition within days, even when provided intermittently. Here we review the detrimental effects of diet and obesity on cognition and the role of inflammatory and circulating factors, compromised blood-brain barrier integrity and gut microbiome changes. We next evaluate evidence for changing risk profiles across life stages (adolescence and ageing) and other populations at risk (e.g. through maternal obesity). Finally, interventions to ameliorate diet-induced cognitive deficits are discussed, including dietary shifts, exercise, and the emerging field of microbiome-targeted therapies. With evidence that poor diet and obesity impair cognition via multiple mechanisms across the human lifespan, the challenge for future research is to identify effective interventions, in addition to diet and exercise, to prevent and ameliorate adverse effects.
Collapse
|
11
|
Orkin JD, Montague MJ, Tejada-Martinez D, de Manuel M, Del Campo J, Cheves Hernandez S, Di Fiore A, Fontsere C, Hodgson JA, Janiak MC, Kuderna LFK, Lizano E, Martin MP, Niimura Y, Perry GH, Valverde CS, Tang J, Warren WC, de Magalhães JP, Kawamura S, Marquès-Bonet T, Krawetz R, Melin AD. The genomics of ecological flexibility, large brains, and long lives in capuchin monkeys revealed with fecalFACS. Proc Natl Acad Sci U S A 2021; 118:e2010632118. [PMID: 33574059 PMCID: PMC7896301 DOI: 10.1073/pnas.2010632118] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Ecological flexibility, extended lifespans, and large brains have long intrigued evolutionary biologists, and comparative genomics offers an efficient and effective tool for generating new insights into the evolution of such traits. Studies of capuchin monkeys are particularly well situated to shed light on the selective pressures and genetic underpinnings of local adaptation to diverse habitats, longevity, and brain development. Distributed widely across Central and South America, they are inventive and extractive foragers, known for their sensorimotor intelligence. Capuchins have among the largest relative brain size of any monkey and a lifespan that exceeds 50 y, despite their small (3 to 5 kg) body size. We assemble and annotate a de novo reference genome for Cebus imitator Through high-depth sequencing of DNA derived from blood, various tissues, and feces via fluorescence-activated cell sorting (fecalFACS) to isolate monkey epithelial cells, we compared genomes of capuchin populations from tropical dry forests and lowland rainforests and identified population divergence in genes involved in water balance, kidney function, and metabolism. Through a comparative genomics approach spanning a wide diversity of mammals, we identified genes under positive selection associated with longevity and brain development. Additionally, we provide a technological advancement in the use of noninvasive genomics for studies of free-ranging mammals. Our intra- and interspecific comparative study of capuchin genomics provides insights into processes underlying local adaptation to diverse and physiologically challenging environments, as well as the molecular basis of brain evolution and longevity.
Collapse
Affiliation(s)
- Joseph D Orkin
- Department of Anthropology and Archaeology, University of Calgary, Calgary, AB T2N 1N4, Canada;
- Institut de Biologia Evolutiva, Universitat Pompeu Fabra, Consejo Superior de Investigaciones Cientificas, 08003 Barcelona, Spain
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T38 6A8, Canada
| | - Michael J Montague
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19146
| | - Daniela Tejada-Martinez
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107
- Doctorado en Ciencias mención Ecología y Evolución, Instituto de Ciencias Ambientales y Evolutivas, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5090000, Chile
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, United Kingdom
| | - Marc de Manuel
- Institut de Biologia Evolutiva, Universitat Pompeu Fabra, Consejo Superior de Investigaciones Cientificas, 08003 Barcelona, Spain
| | - Javier Del Campo
- Institut de Biologia Evolutiva, Universitat Pompeu Fabra, Consejo Superior de Investigaciones Cientificas, 08003 Barcelona, Spain
| | | | - Anthony Di Fiore
- Department of Anthropology and Primate Molecular Ecology and Evolution Laboratory, University of Texas at Austin, Austin, TX 78712
- College of Biological and Environmental Sciences, Universidad San Francisco de Quito, 170901 Cumbayá, Ecuador
| | - Claudia Fontsere
- Institut de Biologia Evolutiva, Universitat Pompeu Fabra, Consejo Superior de Investigaciones Cientificas, 08003 Barcelona, Spain
| | - Jason A Hodgson
- Department of Anthropology, Pennsylvania State University, University Park, PA 16802
- Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, United Kingdom
| | - Mareike C Janiak
- Department of Anthropology and Archaeology, University of Calgary, Calgary, AB T2N 1N4, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T38 6A8, Canada
- School of Science, Engineering and Environment, University of Salford, Salford M5 4WT, United Kingdom
| | - Lukas F K Kuderna
- Institut de Biologia Evolutiva, Universitat Pompeu Fabra, Consejo Superior de Investigaciones Cientificas, 08003 Barcelona, Spain
| | - Esther Lizano
- Institut de Biologia Evolutiva, Universitat Pompeu Fabra, Consejo Superior de Investigaciones Cientificas, 08003 Barcelona, Spain
- Institut Català de Paleontologia Miquel Crusafont, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain
| | - Maria Pia Martin
- Kids Saving the Rainforest Wildlife Rescue Center, 60601 Quepos, Costa Rica
| | - Yoshihito Niimura
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - George H Perry
- Department of Anthropology, Pennsylvania State University, University Park, PA 16802
- Department of Biology, Pennsylvania State University, University Park, PA 16802
| | | | - Jia Tang
- Department of Anthropology and Archaeology, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Wesley C Warren
- Division of Animal Sciences, School of Medicine, University of Missouri, Columbia, MO 65211
| | - João Pedro de Magalhães
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, United Kingdom
| | - Shoji Kawamura
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 277-8562 Chiba, Japan
| | - Tomàs Marquès-Bonet
- Institut de Biologia Evolutiva, Universitat Pompeu Fabra, Consejo Superior de Investigaciones Cientificas, 08003 Barcelona, Spain
- Institut Català de Paleontologia Miquel Crusafont, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain
- Catalan Institution of Research and Advanced Studies, 08010 Barcelona, Spain
- Centro Nacional de Análisis Genómico-Centre for Genomic Regulation, Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Roman Krawetz
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB T38 6A8, Canada
| | - Amanda D Melin
- Department of Anthropology and Archaeology, University of Calgary, Calgary, AB T2N 1N4, Canada;
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T38 6A8, Canada
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB T38 6A8, Canada
| |
Collapse
|
12
|
Palmer D, Fabris F, Doherty A, Freitas AA, de Magalhães JP. Ageing transcriptome meta-analysis reveals similarities and differences between key mammalian tissues. Aging (Albany NY) 2021; 13:3313-3341. [PMID: 33611312 PMCID: PMC7906136 DOI: 10.18632/aging.202648] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 10/29/2020] [Indexed: 12/22/2022]
Abstract
By combining transcriptomic data with other data sources, inferences can be made about functional changes during ageing. Thus, we conducted a meta-analysis on 127 publicly available microarray and RNA-Seq datasets from mice, rats and humans, identifying a transcriptomic signature of ageing across species and tissues. Analyses on subsets of these datasets produced transcriptomic signatures of ageing for brain, heart and muscle. We then applied enrichment analysis and machine learning to functionally describe these signatures, revealing overexpression of immune and stress response genes and underexpression of metabolic and developmental genes. Further analyses revealed little overlap between genes differentially expressed with age in different tissues, despite ageing differentially expressed genes typically being widely expressed across tissues. Additionally we show that the ageing gene expression signatures (particularly the overexpressed signatures) of the whole meta-analysis, brain and muscle tend to include genes that are central in protein-protein interaction networks. We also show that genes underexpressed with age in the brain are highly central in a co-expression network, suggesting that underexpression of these genes may have broad phenotypic consequences. In sum, we show numerous functional similarities between the ageing transcriptomes of these important tissues, along with unique network properties of genes differentially expressed with age in both a protein-protein interaction and co-expression networks.
Collapse
Affiliation(s)
- Daniel Palmer
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK.,Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock, Germany
| | - Fabio Fabris
- School of Computing, University of Kent, Canterbury, Kent, UK
| | - Aoife Doherty
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Alex A Freitas
- School of Computing, University of Kent, Canterbury, Kent, UK
| | - João Pedro de Magalhães
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| |
Collapse
|
13
|
Still Living Better through Chemistry: An Update on Caloric Restriction and Caloric Restriction Mimetics as Tools to Promote Health and Lifespan. Int J Mol Sci 2020; 21:ijms21239220. [PMID: 33287232 PMCID: PMC7729921 DOI: 10.3390/ijms21239220] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 11/30/2020] [Accepted: 11/30/2020] [Indexed: 02/06/2023] Open
Abstract
Caloric restriction (CR), the reduction of caloric intake without inducing malnutrition, is the most reproducible method of extending health and lifespan across numerous organisms, including humans. However, with nearly one-third of the world’s population overweight, it is obvious that caloric restriction approaches are difficult for individuals to achieve. Therefore, identifying compounds that mimic CR is desirable to promote longer, healthier lifespans without the rigors of restricting diet. Many compounds, such as rapamycin (and its derivatives), metformin, or other naturally occurring products in our diets (nutraceuticals), induce CR-like states in laboratory models. An alternative to CR is the removal of specific elements (such as individual amino acids) from the diet. Despite our increasing knowledge of the multitude of CR approaches and CR mimetics, the extent to which these strategies overlap mechanistically remains unclear. Here we provide an update of CR and CR mimetic research, summarizing mechanisms by which these strategies influence genome function required to treat age-related pathologies and identify the molecular fountain of youth.
Collapse
|
14
|
Rubio C, Luna R, Rosiles A, Rubio-Osornio M. Caloric Restriction and Ketogenic Diet Therapy for Epilepsy: A Molecular Approach Involving Wnt Pathway and K ATP Channels. Front Neurol 2020; 11:584298. [PMID: 33250850 PMCID: PMC7676225 DOI: 10.3389/fneur.2020.584298] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 09/28/2020] [Indexed: 12/30/2022] Open
Abstract
Epilepsy is a neurological disorder in which, in many cases, there is poor pharmacological control of seizures. Nevertheless, it may respond beneficially to alternative treatments such as dietary therapy, like the ketogenic diet or caloric restriction. One of the mechanisms of these diets is to produce a hyperpolarization mediated by the adenosine triphosphate (ATP)-sensitive potassium (KATP) channels (KATP channels). An extracellular increase of K+ prevents the release of Ca2+ by inhibiting the signaling of the Wnt pathway and the translocation of β-catenin to the cell nucleus. Wnt ligands hyperpolarize the cells by activating K+ current by Ca2+. Each of the diets described in this paper has in common a lower use of carbohydrates, which leads to biochemical, genetic processes presumed to be involved in the reduction of epileptic seizures. Currently, there is not much information about the genetic processes implicated as well as the possible beneficial effects of diet therapy on epilepsy. In this review, we aim to describe some of the possible genes involved in Wnt pathways, their regulation through the KATP channels which are implicated in each one of the diets, and how they can reduce epileptic seizures at the molecular level.
Collapse
Affiliation(s)
- Carmen Rubio
- Neurophysiology Department, National Institute of Neurology and Neurosurgery, Manuel Velasco Suárez, Mexico City, Mexico
| | - Rudy Luna
- Neurophysiology Department, National Institute of Neurology and Neurosurgery, Manuel Velasco Suárez, Mexico City, Mexico
| | - Artemio Rosiles
- Experimental Laboratory of Neurodegenerative Diseases, National Institute of Neurology and Neurosurgery, Manuel Velasco Suárez, Mexico City, Mexico
| | - Moisés Rubio-Osornio
- Experimental Laboratory of Neurodegenerative Diseases, National Institute of Neurology and Neurosurgery, Manuel Velasco Suárez, Mexico City, Mexico
| |
Collapse
|
15
|
Kruempel JC, Howington MB, Leiser SF. Computational tools for geroscience. TRANSLATIONAL MEDICINE OF AGING 2019; 3:132-143. [PMID: 33241167 PMCID: PMC7685266 DOI: 10.1016/j.tma.2019.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The rapid progress of the past three decades has led the geroscience field near a point where human interventions in aging are plausible. Advances across scientific areas, such as high throughput "-omics" approaches, have led to an exponentially increasing quantity of data available for biogerontologists. To best translate the lifespan and healthspan extending interventions discovered by basic scientists into preventative medicine, it is imperative that the current data are comprehensively utilized to generate testable hypotheses about translational interventions. Building a translational pipeline for geroscience will require both systematic efforts to identify interventions that extend healthspan across taxa and diagnostics that can identify patients who may benefit from interventions prior to the onset of an age-related morbidity. Databases and computational tools that organize and analyze both the wealth of information available on basic biogerontology research and clinical data on aging populations will be critical in developing such a pipeline. Here, we review the current landscape of databases and computational resources available for translational aging research. We discuss key platforms and tools available for aging research, with a focus on how each tool can be used in concert with hypothesis driven experiments to move closer to human interventions in aging.
Collapse
Affiliation(s)
- Joseph C.P. Kruempel
- Molecular & Integrative Physiology Department, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Marshall B. Howington
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Scott F. Leiser
- Molecular & Integrative Physiology Department, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
16
|
Hahn O, Drews LF, Nguyen A, Tatsuta T, Gkioni L, Hendrich O, Zhang Q, Langer T, Pletcher S, Wakelam MJO, Beyer A, Grönke S, Partridge L. A nutritional memory effect counteracts benefits of dietary restriction in old mice. Nat Metab 2019; 1:1059-1073. [PMID: 31742247 PMCID: PMC6861129 DOI: 10.1038/s42255-019-0121-0] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Dietary restriction (DR) during adulthood can greatly extend lifespan and improve metabolic health in diverse species. However, whether DR in mammals is still effective when applied for the first time at old age remains elusive. Here, we report results of a late-life DR switch experiment employing 800 mice, in which 24 months old female mice were switched from ad libitum (AL) to DR or vice versa. Strikingly, the switch from DR-to-AL acutely increases mortality, whereas the switch from AL-to-DR causes only a weak and gradual increase in survival, suggesting a memory of earlier nutrition. RNA-seq profiling in liver, brown (BAT) and white adipose tissue (WAT) demonstrate a largely refractory transcriptional and metabolic response to DR after AL feeding in fat tissue, particularly in WAT, and a proinflammatory signature in aged preadipocytes, which is prevented by chronic DR feeding. Our results provide evidence for a nutritional memory as a limiting factor for DR-induced longevity and metabolic remodeling of WAT in mammals.
Collapse
Affiliation(s)
- Oliver Hahn
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Cellular Networks and Systems Biology, CECAD, University of Cologne, Cologne, Germany
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Lisa F Drews
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - An Nguyen
- Inositide lab, The Babraham Institute, Cambridge, UK
| | - Takashi Tatsuta
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Lisonia Gkioni
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Oliver Hendrich
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Qifeng Zhang
- Inositide lab, The Babraham Institute, Cambridge, UK
| | - Thomas Langer
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Scott Pletcher
- Department of Molecular & Integrative Physiology and the Geriatrics Center, University of Michigan, Ann Arbor, USA
| | | | - Andreas Beyer
- Cellular Networks and Systems Biology, CECAD, University of Cologne, Cologne, Germany.
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.
| | | | - Linda Partridge
- Max Planck Institute for Biology of Ageing, Cologne, Germany.
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London, UK.
| |
Collapse
|
17
|
Identification and Application of Gene Expression Signatures Associated with Lifespan Extension. Cell Metab 2019; 30:573-593.e8. [PMID: 31353263 PMCID: PMC6907080 DOI: 10.1016/j.cmet.2019.06.018] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 04/14/2019] [Accepted: 06/27/2019] [Indexed: 02/06/2023]
Abstract
Several pharmacological, dietary, and genetic interventions that increase mammalian lifespan are known, but general principles of lifespan extension remain unclear. Here, we performed RNA sequencing (RNA-seq) analyses of mice subjected to 8 longevity interventions. We discovered a feminizing effect associated with growth hormone regulation and diminution of sex-related differences. Expanding this analysis to 17 interventions with public data, we observed that many interventions induced similar gene expression changes. We identified hepatic gene signatures associated with lifespan extension across interventions, including upregulation of oxidative phosphorylation and drug metabolism, and showed that perturbed pathways may be shared across tissues. We further applied the discovered longevity signatures to identify new lifespan-extending candidates, such as chronic hypoxia, KU-0063794, and ascorbyl-palmitate. Finally, we developed GENtervention, an app that visualizes associations between gene expression changes and longevity. Overall, this study describes general and specific transcriptomic programs of lifespan extension in mice and provides tools to discover new interventions.
Collapse
|
18
|
Barja G. Towards a unified mechanistic theory of aging. Exp Gerontol 2019; 124:110627. [DOI: 10.1016/j.exger.2019.05.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/08/2019] [Accepted: 05/30/2019] [Indexed: 12/18/2022]
|
19
|
Transcriptional profiling identifies strain-specific effects of caloric restriction and opposite responses in human and mouse white adipose tissue. Aging (Albany NY) 2019; 10:701-746. [PMID: 29708498 PMCID: PMC5940131 DOI: 10.18632/aging.101424] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 04/20/2018] [Indexed: 12/13/2022]
Abstract
Caloric restriction (CR) has been extensively studied in rodents as an intervention to improve lifespan and healthspan. However, effects of CR can be strain- and species-specific. This study used publically available microarray data to analyze expression responses to CR in males from 7 mouse strains (C57BL/6J, BALB/c, C3H, 129, CBA, DBA, B6C3F1) and 4 tissues (epididymal white adipose tissue (eWAT), muscle, heart, cortex). In each tissue, the largest number of strain-specific CR responses was identified with respect to the C57BL/6 strain. In heart and cortex, CR responses in C57BL/6 mice were negatively correlated with responses in other strains. Strain-specific CR responses involved genes associated with olfactory receptors (Olfr1184, Olfr910) and insulin/IGF-1 signaling (Igf1, Irs2). In each strain, CR responses in eWAT were negatively correlated with those in human subcutaneous WAT (scWAT). In human scWAT, CR increased expression of genes associated with stem cell maintenance and vascularization. However, orthologous genes linked to these processes were down-regulated in mouse. These results identify strain-specific CR responses limiting generalization across mouse strains. Differential CR responses in mouse versus human WAT may be due to differences in the depots examined and/or the presence of “thrifty genes” in humans that resist adipose breakdown despite caloric deficit.
Collapse
|
20
|
Tacutu R, Thornton D, Johnson E, Budovsky A, Barardo D, Craig T, Diana E, Lehmann G, Toren D, Wang J, Fraifeld VE, de Magalhães JP. Human Ageing Genomic Resources: new and updated databases. Nucleic Acids Res 2019; 46:D1083-D1090. [PMID: 29121237 PMCID: PMC5753192 DOI: 10.1093/nar/gkx1042] [Citation(s) in RCA: 419] [Impact Index Per Article: 83.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 10/18/2017] [Indexed: 12/17/2022] Open
Abstract
In spite of a growing body of research and data, human ageing remains a poorly understood process. Over 10 years ago we developed the Human Ageing Genomic Resources (HAGR), a collection of databases and tools for studying the biology and genetics of ageing. Here, we present HAGR’s main functionalities, highlighting new additions and improvements. HAGR consists of six core databases: (i) the GenAge database of ageing-related genes, in turn composed of a dataset of >300 human ageing-related genes and a dataset with >2000 genes associated with ageing or longevity in model organisms; (ii) the AnAge database of animal ageing and longevity, featuring >4000 species; (iii) the GenDR database with >200 genes associated with the life-extending effects of dietary restriction; (iv) the LongevityMap database of human genetic association studies of longevity with >500 entries; (v) the DrugAge database with >400 ageing or longevity-associated drugs or compounds; (vi) the CellAge database with >200 genes associated with cell senescence. All our databases are manually curated by experts and regularly updated to ensure a high quality data. Cross-links across our databases and to external resources help researchers locate and integrate relevant information. HAGR is freely available online (http://genomics.senescence.info/).
Collapse
Affiliation(s)
- Robi Tacutu
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK.,Computational Biology of Aging Group, Institute of Biochemistry, Romanian Academy, Bucharest 060031, Romania
| | - Daniel Thornton
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| | - Emily Johnson
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| | - Arie Budovsky
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel.,Judea Regional Research & Development Center, Carmel 90404, Israel
| | - Diogo Barardo
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City 117597, Singapore.,Science Division, Yale-NUS College, Singapore City 138527, Singapore
| | - Thomas Craig
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| | - Eugene Diana
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| | - Gilad Lehmann
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Dmitri Toren
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Jingwei Wang
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| | - Vadim E Fraifeld
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - João P de Magalhães
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| |
Collapse
|
21
|
Komljenovic A, Li H, Sorrentino V, Kutalik Z, Auwerx J, Robinson-Rechavi M. Cross-species functional modules link proteostasis to human normal aging. PLoS Comput Biol 2019; 15:e1007162. [PMID: 31269015 PMCID: PMC6634426 DOI: 10.1371/journal.pcbi.1007162] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 07/16/2019] [Accepted: 06/07/2019] [Indexed: 11/23/2022] Open
Abstract
The evolutionarily conserved nature of the few well-known anti-aging interventions that affect lifespan, such as caloric restriction, suggests that aging-related research in model organisms is directly relevant to human aging. Since human lifespan is a complex trait, a systems-level approach will contribute to a more comprehensive understanding of the underlying aging landscape. Here, we integrate evolutionary and functional information of normal aging across human and model organisms at three levels: gene-level, process-level, and network-level. We identify evolutionarily conserved modules of normal aging across diverse taxa, and notably show proteostasis to be conserved in normal aging. Additionally, we find that mechanisms related to protein quality control network are enriched for genes harboring genetic variants associated with 22 age-related human traits and associated to caloric restriction. These results demonstrate that a systems-level approach, combined with evolutionary conservation, allows the detection of candidate aging genes and pathways relevant to human normal aging.
Collapse
Affiliation(s)
- Andrea Komljenovic
- Department of Ecology and Evolution, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Hao Li
- Laboratory of Integrative Systems Physiology, EPFL, Lausanne, Switzerland
| | | | - Zoltán Kutalik
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- University Center for Primary Care and Public Health, University of Lausanne, Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, EPFL, Lausanne, Switzerland
| | - Marc Robinson-Rechavi
- Department of Ecology and Evolution, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| |
Collapse
|
22
|
The Impact of Caloric Restriction on the Epigenetic Signatures of Aging. Int J Mol Sci 2019; 20:ijms20082022. [PMID: 31022953 PMCID: PMC6515465 DOI: 10.3390/ijms20082022] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 04/19/2019] [Accepted: 04/23/2019] [Indexed: 12/14/2022] Open
Abstract
Aging is characterized by an extensive remodeling of epigenetic patterns, which has been implicated in the physiopathology of age-related diseases. Nutrition plays a significant role in modulating the epigenome, and a growing amount of data indicate that dietary changes can modify the epigenetic marks associated with aging. In this review, we will assess the current advances in the relationship between caloric restriction, a proven anti-aging intervention, and epigenetic signatures of aging. We will specifically discuss the impact of caloric restriction on epigenetic regulation and how some of the favorable effects of caloric restriction on lifespan and healthspan could be mediated by epigenetic modifications.
Collapse
|
23
|
Sahm A, Almaida-Pagán P, Bens M, Mutalipassi M, Lucas-Sánchez A, de Costa Ruiz J, Görlach M, Cellerino A. Analysis of the coding sequences of clownfish reveals molecular convergence in the evolution of lifespan. BMC Evol Biol 2019; 19:89. [PMID: 30975078 PMCID: PMC6460853 DOI: 10.1186/s12862-019-1409-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 03/10/2019] [Indexed: 01/12/2023] Open
Abstract
Background Standard evolutionary theories of aging postulate that reduced extrinsic mortality leads to evolution of longevity. Clownfishes of the genus Amphiprion live in a symbiotic relationship with sea anemones that provide protection from predators. We performed a survey and identified at least two species with a lifespan of over 20 years. Given their small size and ease of captive reproduction, clownfish lend themselves as experimental models of exceptional longevity. To identify genetic correlates of exceptional longevity, we sequenced the transcriptomes of Amphiprion percula and A. clarkii and performed a scan for positively-selected genes (PSGs). Results The PSGs that we identified in the last common clownfish ancestor were compared with PSGs detected in long-lived mole rats and short-lived killifishes revealing convergent evolution in processes such as mitochondrial biogenesis. Among individual genes, the Mitochondrial Transcription Termination Factor 1 (MTERF1), was positively-selected in all three clades, whereas the Glutathione S-Transferase Kappa 1 (GSTK1) was under positive selection in two independent clades. For the latter, homology modelling strongly suggested that positive selection targeted enzymatically important residues. Conclusions These results indicate that specific pathways were recruited in independent lineages evolving an exceptionally extended or shortened lifespan and point to mito-nuclear balance as a key factor. Electronic supplementary material The online version of this article (10.1186/s12862-019-1409-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Arne Sahm
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
| | | | - Martin Bens
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
| | | | | | | | - Matthias Görlach
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
| | - Alessandro Cellerino
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany. .,Bio@SNS, Scuola Normale Superiore, Pisa, Italy.
| |
Collapse
|
24
|
Wahl D, Solon-Biet SM, Wang QP, Wali JA, Pulpitel T, Clark X, Raubenheimer D, Senior AM, Sinclair DA, Cooney GJ, de Cabo R, Cogger VC, Simpson SJ, Le Couteur DG. Comparing the Effects of Low-Protein and High-Carbohydrate Diets and Caloric Restriction on Brain Aging in Mice. Cell Rep 2018; 25:2234-2243.e6. [PMID: 30463018 PMCID: PMC6296764 DOI: 10.1016/j.celrep.2018.10.070] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 04/27/2018] [Accepted: 10/19/2018] [Indexed: 12/15/2022] Open
Abstract
Calorie restriction (CR) increases lifespan and improves brain health in mice. Ad libitum low-protein, high-carbohydrate (LPHC) diets also extend lifespan, but it is not known whether they are beneficial for brain health. We compared hippocampus biology and memory in mice subjected to 20% CR or provided ad libitum access to one of three LPHC diets or to a control diet. Patterns of RNA expression in the hippocampus of 15-month-old mice were similar between mice fed CR and LPHC diets when we looked at genes associated with longevity, cytokines, and dendrite morphogenesis. Nutrient-sensing proteins, including SIRT1, mTOR, and PGC1α, were also influenced by diet; however, the effects varied by sex. CR and LPHC diets were associated with increased dendritic spines in dentate gyrus neurons. Mice fed CR and LPHC diets had modest improvements in the Barnes maze and novel object recognition. LPHC diets recapitulate some of the benefits of CR on brain aging.
Collapse
Affiliation(s)
- Devin Wahl
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; Aging and Alzheimers Institute, ANZAC Research Institute, Centre for Education and Research on Ageing, Concord, NSW 2139, Australia
| | | | - Qiao-Ping Wang
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Jibran A Wali
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Tamara Pulpitel
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Ximonie Clark
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - David Raubenheimer
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Alistair M Senior
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; School of Mathematics and Statistics, The University of Sydney, NSW 2006, Australia
| | - David A Sinclair
- Department of Genetics, Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA 02115, USA; Department of Pharmacology, School of Medical Sciences, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Gregory J Cooney
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Victoria C Cogger
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; Aging and Alzheimers Institute, ANZAC Research Institute, Centre for Education and Research on Ageing, Concord, NSW 2139, Australia
| | - Stephen J Simpson
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia.
| | - David G Le Couteur
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; Aging and Alzheimers Institute, ANZAC Research Institute, Centre for Education and Research on Ageing, Concord, NSW 2139, Australia.
| |
Collapse
|
25
|
Gokarn R, Solon-Biet SM, Cogger VC, Cooney GJ, Wahl D, McMahon AC, Mitchell JR, Mitchell SJ, Hine C, de Cabo R, Raubenheimer D, Simpson SJ, Le Couteur DG. Long-term Dietary Macronutrients and Hepatic Gene Expression in Aging Mice. J Gerontol A Biol Sci Med Sci 2018; 73:1618-1625. [PMID: 29688281 PMCID: PMC6454416 DOI: 10.1093/gerona/gly065] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Indexed: 12/13/2022] Open
Abstract
Nutrition influences both hepatic function and aging, but mechanisms are poorly understood. Here, the effects of lifelong, ad libitum-fed diets varying in macronutrients and energy on hepatic gene expression were studied. Gene expression was measured using Affymetrix mouse arrays in livers of 46 mice aged 15 months fed one of 25 diets varying in protein, carbohydrates, fat, and energy density from 3 weeks of age. Gene expression was almost entirely influenced by protein intake. Carbohydrate and fat intake had few effects on gene expression compared with protein. Pathways and processes associated with protein intake included those involved with mitochondrial function, metabolic signaling (PI3K-Akt, AMPK, mTOR) and metabolism of protein and amino acids. Protein intake had variable effects on genes associated with regulation of longevity and influenced by caloric restriction. Among the genes of interest with expression that were significantly associated with protein intake are Cth, Gls2, Igf1, and Nnmt, which were increased with higher protein intake, and Igf2bp2, Fgf21, Prkab2, and Mtor, which were increased with lower protein intake. Dietary protein has a powerful impact on hepatic gene expression in older mice, with some overlap with genes previously reported to be involved with regulation of longevity or caloric restriction.
Collapse
Affiliation(s)
- Rahul Gokarn
- Charles Perkins Centre, University of Sydney, New South Wales, Australia
- Aging and Alzheimers Institute (AAAI), New South Wales, Australia
- Centre for Education and Research on Ageing (CERA), New South Wales, Australia
- ANZAC Research Institute, Concord Hospital, New South Wales, Australia
| | - Samantha M Solon-Biet
- Charles Perkins Centre, University of Sydney, New South Wales, Australia
- Aging and Alzheimers Institute (AAAI), New South Wales, Australia
- Centre for Education and Research on Ageing (CERA), New South Wales, Australia
- ANZAC Research Institute, Concord Hospital, New South Wales, Australia
| | - Victoria C Cogger
- Charles Perkins Centre, University of Sydney, New South Wales, Australia
- Aging and Alzheimers Institute (AAAI), New South Wales, Australia
- Centre for Education and Research on Ageing (CERA), New South Wales, Australia
- ANZAC Research Institute, Concord Hospital, New South Wales, Australia
| | - Gregory J Cooney
- Charles Perkins Centre, University of Sydney, New South Wales, Australia
| | - Devin Wahl
- Charles Perkins Centre, University of Sydney, New South Wales, Australia
- Aging and Alzheimers Institute (AAAI), New South Wales, Australia
- Centre for Education and Research on Ageing (CERA), New South Wales, Australia
- ANZAC Research Institute, Concord Hospital, New South Wales, Australia
| | - Aisling C McMahon
- Aging and Alzheimers Institute (AAAI), New South Wales, Australia
- Centre for Education and Research on Ageing (CERA), New South Wales, Australia
- ANZAC Research Institute, Concord Hospital, New South Wales, Australia
| | - James R Mitchell
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, Massachusetts
| | - Sarah J Mitchell
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, Massachusetts
| | - Christopher Hine
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, Massachusetts
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - David Raubenheimer
- Charles Perkins Centre, University of Sydney, New South Wales, Australia
| | - Stephen J Simpson
- Charles Perkins Centre, University of Sydney, New South Wales, Australia
| | - David G Le Couteur
- Charles Perkins Centre, University of Sydney, New South Wales, Australia
- Aging and Alzheimers Institute (AAAI), New South Wales, Australia
- Centre for Education and Research on Ageing (CERA), New South Wales, Australia
- ANZAC Research Institute, Concord Hospital, New South Wales, Australia
| |
Collapse
|
26
|
Hahn O, Stubbs TM, Reik W, Grönke S, Beyer A, Partridge L. Hepatic gene body hypermethylation is a shared epigenetic signature of murine longevity. PLoS Genet 2018; 14:e1007766. [PMID: 30462643 PMCID: PMC6281273 DOI: 10.1371/journal.pgen.1007766] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 12/05/2018] [Accepted: 11/08/2018] [Indexed: 12/30/2022] Open
Abstract
Dietary, pharmacological and genetic interventions can extend health- and lifespan in diverse mammalian species. DNA methylation has been implicated in mediating the beneficial effects of these interventions; methylation patterns deteriorate during ageing, and this is prevented by lifespan-extending interventions. However, whether these interventions also actively shape the epigenome, and whether such epigenetic reprogramming contributes to improved health at old age, remains underexplored. We analysed published, whole-genome, BS-seq data sets from mouse liver to explore DNA methylation patterns in aged mice in response to three lifespan-extending interventions: dietary restriction (DR), reduced TOR signaling (rapamycin), and reduced growth (Ames dwarf mice). Dwarf mice show enhanced DNA hypermethylation in the body of key genes in lipid biosynthesis, cell proliferation and somatotropic signaling, which strongly correlates with the pattern of transcriptional repression. Remarkably, DR causes a similar hypermethylation in lipid biosynthesis genes, while rapamycin treatment increases methylation signatures in genes coding for growth factor and growth hormone receptors. Shared changes of DNA methylation were restricted to hypermethylated regions, and they were not merely a consequence of slowed ageing, thus suggesting an active mechanism driving their formation. By comparing the overlap in ageing-independent hypermethylated patterns between all three interventions, we identified four regions, which, independent of genetic background or gender, may serve as novel biomarkers for longevity-extending interventions. In summary, we identified gene body hypermethylation as a novel and partly conserved signature of lifespan-extending interventions in mouse, highlighting epigenetic reprogramming as a possible intervention to improve health at old age.
Collapse
Affiliation(s)
- Oliver Hahn
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Cellular Networks and Systems Biology, CECAD, University of Cologne, Cologne, Germany
| | - Thomas M. Stubbs
- Epigenetics Programme, The Babraham Institute, Cambridge, United Kingdom
| | - Wolf Reik
- Epigenetics Programme, The Babraham Institute, Cambridge, United Kingdom
- The Wellcome Trust Sanger Institute, Cambridge, United Kingdom
| | | | - Andreas Beyer
- Cellular Networks and Systems Biology, CECAD, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Linda Partridge
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London, United Kingdom
| |
Collapse
|
27
|
Azzouz-Olden F, Hunt A, DeGrandi-Hoffman G. Transcriptional response of honey bee (Apis mellifera) to differential nutritional status and Nosema infection. BMC Genomics 2018; 19:628. [PMID: 30134827 PMCID: PMC6106827 DOI: 10.1186/s12864-018-5007-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 08/13/2018] [Indexed: 12/29/2022] Open
Abstract
Background Bees are confronting several environmental challenges, including the intermingled effects of malnutrition and disease. Intuitively, pollen is the healthiest nutritional choice, however, commercial substitutes, such as Bee-Pro and MegaBee, are widely used. Herein we examined how feeding natural and artificial diets shapes transcription in the abdomen of the honey bee, and how transcription shifts in combination with Nosema parasitism. Results Gene ontology enrichment revealed that, compared with poor diet (carbohydrates [C]), bees fed pollen (P > C), Bee-Pro (B > C), and MegaBee (M > C) showed a broad upregulation of metabolic processes, especially lipids; however, pollen feeding promoted more functions, and superior proteolysis. The superiority of the pollen diet was also evident through the remarkable overexpression of vitellogenin in bees fed pollen instead of MegaBee or Bee-Pro. Upregulation of bioprocesses under carbohydrates feeding compared to pollen (C > P) provided a clear poor nutritional status, uncovering stark expression changes that were slight or absent relatively to Bee-Pro (C > B) or MegaBee (C > M). Poor diet feeding (C > P) induced starvation response genes and hippo signaling pathway, while it repressed growth through different mechanisms. Carbohydrate feeding (C > P) also elicited ‘adult behavior’, and developmental processes suggesting transition to foraging. Finally, it altered the ‘circadian rhythm’, reflecting the role of this mechanism in the adaptation to nutritional stress in mammals. Nosema-infected bees fed pollen compared to carbohydrates (PN > CN) upheld certain bioprocesses of uninfected bees (P > C). Poor nutritional status was more apparent against pollen (CN > PN) than Bee-Pro (CN > BN) or MegaBee (CN > MN). Nosema accentuated the effects of malnutrition since more starvation-response genes and stress response mechanisms were upregulated in CN > PN compared to C > P. The bioprocess ‘Macromolecular complex assembly’ was also enriched in CN > PN, and involved genes associated with human HIV and/or influenza, thus providing potential candidates for bee-Nosema interactions. Finally, the enzyme Duox emerged as essential for guts defense in bees, similarly to Drosophila. Conclusions These results provide evidence of the superior nutritional status of bees fed pollen instead of artificial substitutes in terms of overall health, even in the presence of a pathogen. Electronic supplementary material The online version of this article (10.1186/s12864-018-5007-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Arthur Hunt
- Department of Plant and Soil Sciences, University of Kentucky, Lexington, KY, 40546, USA
| | | |
Collapse
|
28
|
Ma S, Avanesov AS, Porter E, Lee BC, Mariotti M, Zemskaya N, Guigo R, Moskalev AA, Gladyshev VN. Comparative transcriptomics across 14 Drosophila species reveals signatures of longevity. Aging Cell 2018; 17:e12740. [PMID: 29671950 PMCID: PMC6052463 DOI: 10.1111/acel.12740] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2018] [Indexed: 12/14/2022] Open
Abstract
Lifespan varies dramatically among species, but the biological basis is not well understood. Previous studies in model organisms revealed the importance of nutrient sensing, mTOR, NAD/sirtuins, and insulin/IGF1 signaling in lifespan control. By studying life-history traits and transcriptomes of 14 Drosophila species differing more than sixfold in lifespan, we explored expression divergence and identified genes and processes that correlate with longevity. These longevity signatures suggested that longer-lived flies upregulate fatty acid metabolism, downregulate neuronal system development and activin signaling, and alter dynamics of RNA splicing. Interestingly, these gene expression patterns resembled those of flies under dietary restriction and several other lifespan-extending interventions, although on the individual gene level, there was no significant overlap with genes previously reported to have lifespan-extension effects. We experimentally tested the lifespan regulation potential of several candidate genes and found no consistent effects, suggesting that individual genes generally do not explain the observed longevity patterns. Instead, it appears that lifespan regulation across species is modulated by complex relationships at the system level represented by global gene expression.
Collapse
Affiliation(s)
- Siming Ma
- Division of GeneticsDepartment of MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
- Genome Institute of SingaporeA*STARSingapore CitySingapore
| | - Andrei S. Avanesov
- Division of GeneticsDepartment of MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Emily Porter
- Division of GeneticsDepartment of MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Byung Cheon Lee
- Division of GeneticsDepartment of MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
- College of Life Sciences and BiotechnologyKorea UniversitySeoulSouth Korea
| | - Marco Mariotti
- Division of GeneticsDepartment of MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
- Bioinformatics and Genomics ProgramCentre for Genomic Regulation and Universitat Pompeu FabraBarcelonaSpain
| | - Nadezhda Zemskaya
- Institute of BiologyKomi Science CenterRussian Academy of SciencesSyktyvkarRussia
| | - Roderic Guigo
- Bioinformatics and Genomics ProgramCentre for Genomic Regulation and Universitat Pompeu FabraBarcelonaSpain
| | - Alexey A. Moskalev
- Institute of BiologyKomi Science CenterRussian Academy of SciencesSyktyvkarRussia
- Moscow Institute of Physics and TechnologyDolgoprudny, Moscow RegionRussia
- Engelhardt Institute of Molecular BiologyRussian Academy of SciencesMoscowRussia
| | - Vadim N. Gladyshev
- Division of GeneticsDepartment of MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
- Belozersky Institute of Physico‐Chemical BiologyMoscow State UniversityMoscowRussia
| |
Collapse
|
29
|
Abstract
Caloric restriction (CR) extends lifespan and delays the onset of age-related disorders in diverse species. Metabolic regulatory pathways have been implicated in the mechanisms of CR, but the molecular details have not been elucidated. Here, we show that CR engages RNA processing of genes associated with a highly integrated reprogramming of hepatic metabolism. We conducted molecular profiling of liver biopsies collected from adult male rhesus monkeys (Macaca mulatta) at baseline and after 2 years on control or CR (30% restricted) diet. Quantitation of over 20,000 molecules from the hepatic transcriptome, proteome, and metabolome indicated that metabolism and RNA processing are major features of the response to CR. Predictive models identified lipid, branched-chain amino acid, and short-chain carbon metabolic pathways, with alternate transcript use for over half of the genes in the CR network. We conclude that RNA-based mechanisms are central to the CR response and integral in metabolic reprogramming.
Collapse
|
30
|
Abstract
Ageing leads to dramatic changes in the physiology of many different tissues resulting in a spectrum of pathology. Nonetheless, many lines of evidence suggest that ageing is driven by highly conserved cell intrinsic processes, and a set of unifying hallmarks of ageing has been defined. Here, we survey reports of age-linked changes in basal gene expression across eukaryotes from yeast to human and identify six gene expression hallmarks of cellular ageing: downregulation of genes encoding mitochondrial proteins; downregulation of the protein synthesis machinery; dysregulation of immune system genes; reduced growth factor signalling; constitutive responses to stress and DNA damage; dysregulation of gene expression and mRNA processing. These encompass widely reported features of ageing such as increased senescence and inflammation, reduced electron transport chain activity and reduced ribosome synthesis, but also reveal a surprising lack of gene expression responses to known age-linked cellular stresses. We discuss how the existence of conserved transcriptomic hallmarks relates to genome-wide epigenetic differences underlying ageing clocks, and how the changing transcriptome results in proteomic alterations where data is available and to variations in cell physiology characteristic of ageing. Identification of gene expression events that occur during ageing across distant organisms should be informative as to conserved underlying mechanisms of ageing, and provide additional biomarkers to assess the effects of diet and other environmental factors on the rate of ageing.
Collapse
Affiliation(s)
- Stephen Frenk
- Department of Genetics, University of North Carolina, Chapel Hill, NC, 27599-3280, USA
| | | |
Collapse
|
31
|
Bettedi L, Foukas LC. Growth factor, energy and nutrient sensing signalling pathways in metabolic ageing. Biogerontology 2017; 18:913-929. [PMID: 28795262 PMCID: PMC5684302 DOI: 10.1007/s10522-017-9724-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 07/21/2017] [Indexed: 01/24/2023]
Abstract
The field of the biology of ageing has received increasing attention from a biomedical point of view over the past decades. The main reason has been the realisation that increases in human population life expectancy are accompanied by late onset diseases. Indeed, ageing is the most important risk factor for a number of neoplastic, neurodegenerative and metabolic pathologies. Advances in the knowledge of the genetics of ageing, mainly through research in model organisms, have implicated various cellular processes and the respective signalling pathways that regulate them in cellular and organismal ageing. Associated with ageing is a dysregulation of metabolic homeostasis usually manifested as age-related obesity, diminished insulin sensitivity and impaired glucose and lipid homeostasis. Metabolic deterioration contributes to the ageing phenotype and metabolic pathologies are thought to be one of the main factors limiting the potential for lifespan extension. Great efforts have been directed towards identifying pharmacological interventions with the potential to improve healthspan and a number of natural and synthetic compounds have shown promise in achieving beneficial metabolic effects.
Collapse
Affiliation(s)
- Lucia Bettedi
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, London, UK
- Cell Biology and Neurobiology Branch, National Institutes of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Lazaros C Foukas
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, London, UK.
| |
Collapse
|
32
|
The effects of graded levels of calorie restriction: VII. Topological rearrangement of hypothalamic aging networks. Aging (Albany NY) 2017; 8:917-32. [PMID: 27115072 PMCID: PMC4931844 DOI: 10.18632/aging.100944] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 03/31/2016] [Indexed: 12/31/2022]
Abstract
Connectivity in a gene-gene network declines with age, typically within gene clusters. We explored the effect of short-term (3 months) graded calorie restriction (CR) (up to 40 %) on network structure of aging-associated genes in the murine hypothalamus by using conditional mutual information. The networks showed a topological rearrangement when exposed to graded CR with a higher relative within cluster connectivity at 40CR. We observed changes in gene centrality concordant with changes in CR level, with Ppargc1a, and Ppt1 having increased centrality and Etfdh, Traf3 and Abcc1 decreased centrality as CR increased. This change in gene centrality in a graded manner with CR, occurred in the absence of parallel changes in gene expression levels. This study emphasizes the importance of augmenting traditional differential gene expression analyses to better understand structural changes in the transcriptome. Overall our results suggested that CR induced changes in centrality of biological relevant genes that play an important role in preventing the age-associated loss of network integrity irrespective of their gene expression levels.
Collapse
|
33
|
Bustos V, Partridge L. Good Ol' Fat: Links between Lipid Signaling and Longevity. Trends Biochem Sci 2017; 42:812-823. [PMID: 28802547 DOI: 10.1016/j.tibs.2017.07.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 07/07/2017] [Accepted: 07/13/2017] [Indexed: 12/13/2022]
Abstract
Aging is the single greatest risk factor for the development of disease. Understanding the biological molecules and mechanisms that modulate aging is therefore critical for the development of health-maximizing interventions for older people. The effect of fats on longevity has traditionally been disregarded as purely detrimental. However, new studies are starting to uncover the possible beneficial effects of lipids working as signaling molecules on health and longevity. These studies highlight the complex links between aging and lipid signaling. In this review we summarize accumulating evidence that points to changes in lipid metabolism, and in particular lipid signaling, as an underlying mechanism for healthy aging.
Collapse
Affiliation(s)
- Victor Bustos
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Strasse 9b, 50931, Cologne, Germany
| | - Linda Partridge
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Strasse 9b, 50931, Cologne, Germany; Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, Darwin Building, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
34
|
Barger JL, Vann JM, Cray NL, Pugh TD, Mastaloudis A, Hester SN, Wood SM, Newton MA, Weindruch R, Prolla TA. Identification of tissue-specific transcriptional markers of caloric restriction in the mouse and their use to evaluate caloric restriction mimetics. Aging Cell 2017; 16:750-760. [PMID: 28556428 PMCID: PMC5506434 DOI: 10.1111/acel.12608] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2017] [Indexed: 12/28/2022] Open
Abstract
Caloric restriction (CR) without malnutrition has been shown to retard several aspects of the aging process and to extend lifespan in different species. There is strong interest in the identification of CR mimetics (CRMs), compounds that mimic the beneficial effects of CR on lifespan and healthspan without restriction of energy intake. Identification of CRMs in mammals is currently inefficient due to the lack of screening tools. We have performed whole‐genome transcriptional profiling of CR in seven mouse strains (C3H/HeJ, CBA/J, DBA/2J, B6C3F1/J, 129S1/SvImJ, C57BL/6J, and BALB/cJ) in white adipose tissue (WAT), gastrocnemius muscle, heart, and brain neocortex. This analysis has identified tissue‐specific panels of genes that change in expression in multiple mouse strains with CR. We validated a subset of genes with qPCR and used these to evaluate the potential CRMs bezafibrate, pioglitazone, metformin, resveratrol, quercetin, 2,4‐dinitrophenol, and L‐carnitine when fed to C57BL/6J 2‐month‐old mice for 3 months. Compounds were also evaluated for their ability to modulate previously characterized biomarkers of CR, including mitochondrial enzymes citrate synthase and SIRT3, plasma inflammatory cytokines TNF‐α and IFN‐γ, glycated hemoglobin (HbA1c) levels and adipocyte size. Pioglitazone, a PPAR‐γ agonist, and L‐carnitine, an amino acid involved in lipid metabolism, displayed the strongest effects on both the novel transcriptional markers of CR and the additional CR biomarkers tested. Our findings provide panels of tissue‐specific transcriptional markers of CR that can be used to identify novel CRMs, and also represent the first comparative molecular analysis of several potential CRMs in multiple tissues in mammals.
Collapse
Affiliation(s)
| | | | | | | | | | - Shelly N. Hester
- Center for Anti-Aging Research; NSE Products, Inc.; Provo UT USA
| | - Steven M. Wood
- Center for Anti-Aging Research; NSE Products, Inc.; Provo UT USA
| | - Michael A. Newton
- Departments of Statistics and of Biostatistics and Medical Informatics; University of Wisconsin; Madison WI USA
| | - Richard Weindruch
- LifeGen Technologies LLC; Madison WI USA
- Department of Medicine; SMPH; University of Wisconsin; Madison WI USA
- Geriatric Research, Education and Clinical Center; William S. Middleton Memorial Veterans Hospital; Madison WI USA
| | - Tomas A. Prolla
- LifeGen Technologies LLC; Madison WI USA
- Departments of Genetics and Medical Genetics; University of Wisconsin; Madison WI USA
| |
Collapse
|
35
|
de Magalhães JP, Stevens M, Thornton D. The Business of Anti-Aging Science. Trends Biotechnol 2017; 35:1062-1073. [PMID: 28778607 DOI: 10.1016/j.tibtech.2017.07.004] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 06/18/2017] [Accepted: 07/06/2017] [Indexed: 12/16/2022]
Abstract
Age-related conditions are the leading causes of death and health-care costs. Reducing the rate of aging would have enormous medical and financial benefits. Myriad genes and pathways are known to regulate aging in model organisms, fostering a new crop of anti-aging companies. Approaches range from drug discovery efforts to big-data methods and direct-to-consumer (DTC) strategies. Challenges and pitfalls of commercialization include reliance on findings from short-lived model organisms, poor biological understanding of aging, and hurdles in performing clinical trials for aging. A large number of potential aging-associated interventions and targets exist, but given the long validation times only a small fraction can be explored for clinical applications. If even one company succeeds, however, the impact will be huge.
Collapse
Affiliation(s)
- João Pedro de Magalhães
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK; Joint first authors.
| | - Michael Stevens
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK; Joint first authors
| | - Daniel Thornton
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| |
Collapse
|
36
|
Green CL, Mitchell SE, Derous D, Wang Y, Chen L, Han JDJ, Promislow DEL, Lusseau D, Douglas A, Speakman JR. The effects of graded levels of calorie restriction: IX. Global metabolomic screen reveals modulation of carnitines, sphingolipids and bile acids in the liver of C57BL/6 mice. Aging Cell 2017; 16:529-540. [PMID: 28139067 PMCID: PMC5418186 DOI: 10.1111/acel.12570] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2016] [Indexed: 12/12/2022] Open
Abstract
Calorie restriction (CR) remains the most robust intervention to extend lifespan and improve health span. Using a global mass spectrometry-based metabolomic approach, we identified 193 metabolites that were significantly differentially expressed (SDE) in the livers of C57BL/6 mice, fed graded levels of CR (10, 20, 30 and 40% CR) compared to mice fed ad libitum for 12 h a day. The differential expression of metabolites also varied with the different feeding groups. Pathway analysis revealed that graded CR had an impact on carnitine synthesis and the carnitine shuttle pathway, sphingosine-1-phosphate (S1P) signalling and methionine metabolism. S1P, sphingomyelin and L-carnitine were negatively correlated with body mass, leptin, insulin-like growth factor- 1 (IGF-1) and major urinary proteins (MUPs). In addition, metabolites which showed a graded effect, such as ceramide, S1P, taurocholic acid and L-carnitine, responded in the opposite direction to previously observed age-related changes. We suggest that the modulation of this set of metabolites may improve liver processes involved in energy release from fatty acids. S1P also negatively correlated with catalase activity and body temperature, and positively correlated with food anticipatory activity. Injecting mice with S1P or an S1P receptor 1 agonist did not precipitate changes in body temperature, physical activity or food intake suggesting that these correlations were not causal relationships.
Collapse
Affiliation(s)
- Cara L. Green
- Institute of Biological and Environmental Sciences; University of Aberdeen; Aberdeen UK
| | - Sharon E. Mitchell
- Institute of Biological and Environmental Sciences; University of Aberdeen; Aberdeen UK
| | - Davina Derous
- Institute of Biological and Environmental Sciences; University of Aberdeen; Aberdeen UK
| | - Yingchun Wang
- State Key Laboratory of Molecular Developmental Biology; Institute of Genetics and Developmental Biology; Chinese Academy of Sciences; Chaoyang Beijing China
| | - Luonan Chen
- Key Laboratory of Systems Biology, Innovation Center for Cell Signaling Network; Institute of Biochemistry and Cell Biology; Shanghai Institute of Biological Sciences; Chinese Academy of Sciences; Shanghai China
| | - Jing-Dong J. Han
- Key Laboratory of Computational Biology; Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology; Shanghai Institutes for Biological Sciences; Chinese Academy of Sciences; Shanghai China
| | - Daniel E. L. Promislow
- Department of Pathology and Department of Biology; University of Washington; Seattle WA USA
| | - David Lusseau
- Institute of Biological and Environmental Sciences; University of Aberdeen; Aberdeen UK
| | - Alex Douglas
- Institute of Biological and Environmental Sciences; University of Aberdeen; Aberdeen UK
| | - John R. Speakman
- Institute of Biological and Environmental Sciences; University of Aberdeen; Aberdeen UK
- State Key Laboratory of Molecular Developmental Biology; Institute of Genetics and Developmental Biology; Chinese Academy of Sciences; Chaoyang Beijing China
| |
Collapse
|
37
|
Hahn O, Grönke S, Stubbs TM, Ficz G, Hendrich O, Krueger F, Andrews S, Zhang Q, Wakelam MJ, Beyer A, Reik W, Partridge L. Dietary restriction protects from age-associated DNA methylation and induces epigenetic reprogramming of lipid metabolism. Genome Biol 2017; 18:56. [PMID: 28351387 PMCID: PMC5370449 DOI: 10.1186/s13059-017-1187-1] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 03/02/2017] [Indexed: 12/11/2022] Open
Abstract
Background Dietary restriction (DR), a reduction in food intake without malnutrition, increases most aspects of health during aging and extends lifespan in diverse species, including rodents. However, the mechanisms by which DR interacts with the aging process to improve health in old age are poorly understood. DNA methylation could play an important role in mediating the effects of DR because it is sensitive to the effects of nutrition and can affect gene expression memory over time. Results Here, we profile genome-wide changes in DNA methylation, gene expression and lipidomics in response to DR and aging in female mouse liver. DR is generally strongly protective against age-related changes in DNA methylation. During aging with DR, DNA methylation becomes targeted to gene bodies and is associated with reduced gene expression, particularly of genes involved in lipid metabolism. The lipid profile of the livers of DR mice is correspondingly shifted towards lowered triglyceride content and shorter chain length of triglyceride-associated fatty acids, and these effects become more pronounced with age. Conclusions Our results indicate that DR remodels genome-wide patterns of DNA methylation so that age-related changes are profoundly delayed, while changes at loci involved in lipid metabolism affect gene expression and the resulting lipid profile. Electronic supplementary material The online version of this article (doi:10.1186/s13059-017-1187-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Oliver Hahn
- Max Planck Institute for Biology of Ageing, 50931, Cologne, Germany.,Cellular Networks and Systems Biology, CECAD, University of Cologne, Joseph-Stelzmann-Str. 26, Cologne, 50931, Germany
| | - Sebastian Grönke
- Max Planck Institute for Biology of Ageing, 50931, Cologne, Germany
| | - Thomas M Stubbs
- Epigenetics Programme, The Babraham Institute, Cambridge, CB22 3AT, UK
| | - Gabriella Ficz
- Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Oliver Hendrich
- Max Planck Institute for Biology of Ageing, 50931, Cologne, Germany
| | - Felix Krueger
- Bioinformatics Group, The Babraham Institute, Cambridge, CB22 3AT, UK
| | - Simon Andrews
- Bioinformatics Group, The Babraham Institute, Cambridge, CB22 3AT, UK
| | - Qifeng Zhang
- Inositide Lab, The Babraham Institute, Cambridge, CB22 3AT, UK
| | | | - Andreas Beyer
- Cellular Networks and Systems Biology, CECAD, University of Cologne, Joseph-Stelzmann-Str. 26, Cologne, 50931, Germany. .,Center for Molecular Medicine Cologne, University of Cologne, Cologne, 50931, Germany.
| | - Wolf Reik
- Epigenetics Programme, The Babraham Institute, Cambridge, CB22 3AT, UK. .,The Wellcome Trust Sanger Institute, Cambridge, CB10 1SA, UK.
| | - Linda Partridge
- Max Planck Institute for Biology of Ageing, 50931, Cologne, Germany. .,Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
38
|
Gillespie ZE, Pickering J, Eskiw CH. Better Living through Chemistry: Caloric Restriction (CR) and CR Mimetics Alter Genome Function to Promote Increased Health and Lifespan. Front Genet 2016; 7:142. [PMID: 27588026 PMCID: PMC4988992 DOI: 10.3389/fgene.2016.00142] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 07/21/2016] [Indexed: 12/19/2022] Open
Abstract
Caloric restriction (CR), defined as decreased nutrient intake without causing malnutrition, has been documented to increase both health and lifespan across numerous organisms, including humans. Many drugs and other compounds naturally occurring in our diet (nutraceuticals) have been postulated to act as mimetics of caloric restriction, leading to a wave of research investigating the efficacy of these compounds in preventing age-related diseases and promoting healthier, longer lifespans. Although well studied at the biochemical level, there are still many unanswered questions about how CR and CR mimetics impact genome function and structure. Here we discuss how genome function and structure are influenced by CR and potential CR mimetics, including changes in gene expression profiles and epigenetic modifications and their potential to identify the genetic fountain of youth.
Collapse
Affiliation(s)
- Zoe E Gillespie
- Department of Food and Bioproduct Sciences, University of Saskatchewan Saskatoon, SK, Canada
| | - Joshua Pickering
- Department of Biochemistry, University of Saskatchewan Saskatoon, SK, Canada
| | - Christopher H Eskiw
- Department of Food and Bioproduct Sciences, University of SaskatchewanSaskatoon, SK, Canada; Department of Biochemistry, University of SaskatchewanSaskatoon, SK, Canada
| |
Collapse
|
39
|
Suchiang K, Sharma R. Age-dependent modulation of fasting and long-term dietary restriction on acetylcholinesterase in non-neuronal tissues of mice. Mol Cell Biochem 2016; 419:135-45. [PMID: 27379505 DOI: 10.1007/s11010-016-2757-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 06/21/2016] [Indexed: 12/18/2022]
Abstract
Dietary restriction (DR) without malnutrition is a robust intervention that extends lifespan and slows the onset of nervous system deficit and age-related diseases in diverse organisms. Acetylcholinesterase (AChE), a thoroughly studied enzyme better known for hydrolyzing acetylcholine (ACh) in neuronal tissues, has recently been linked with multiple unrelated biological functions in different non-neuronal tissues. In the present study, the activity and protein expression level of AChE in liver, heart, and kidney of young (1 month), adult (6 month), and aged (18 month) mice were investigated. We also studied age- and tissue-specific changes in AChE activity and protein expression level after the mice were subjected to 24-h fasting and long-term DR. Our results showed that AChE activity and protein expression in kidney and heart of aged mice decreased significantly in comparison with young mice. On the contrary, long-term DR decreases the AChE activity and the protein expression level in all tissues irrespective of ages studied. We summarized that changes in AChE with age in different tissues studied reflects its different roles at different phases of an organism's life. Conversely, the cumulative modulation manifested in the form of lowering AChE by long-term DR may prevent the futile synthesis and accumulation of unwanted AChE besides the added compensatory benefit of enhanced ACh availability needed during the period of starvation. This, in turn, may help in preventing the declining homeostatic roles of this important neurotransmitter in different tissues.
Collapse
Affiliation(s)
- Kitlangki Suchiang
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, 605 014, India.
| | - Ramesh Sharma
- Department of Biochemistry, North-Eastern Hill University, Shillong, Meghalaya, 793 022, India
| |
Collapse
|
40
|
Van Bussel IPG, Jolink-Stoppelenburg A, De Groot CPGM, Müller MR, Afman LA. Differences in genome-wide gene expression response in peripheral blood mononuclear cells between young and old men upon caloric restriction. GENES AND NUTRITION 2016; 11:13. [PMID: 27551314 PMCID: PMC4968441 DOI: 10.1186/s12263-016-0528-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 04/19/2016] [Indexed: 12/02/2022]
Abstract
Background Caloric restriction (CR) is considered to increase lifespan and to prevent various age-related diseases in different nonhuman organisms. Only a limited number of CR studies have been performed on humans, and results put CR as a beneficial tool to decrease risk factors in several age-related diseases. The question remains at what age CR should be implemented to be most effective with respect to healthy aging. The aim of our study was to elucidate the role of age in the transcriptional response to a completely controlled 30 % CR diet on immune cells, as immune response is affected during aging. Ten healthy young men, aged 20–28, and nine healthy old men, aged 64–85, were subjected to a 2-week weight maintenance diet, followed by 3 weeks of 30 % CR. Before and after 30 % CR, the whole genome gene expression in peripheral blood mononuclear cells (PBMCs) was assessed. Results Expression of 554 genes showed a different response between young and old men upon CR. Gene set enrichment analysis revealed a downregulation of gene sets involved in the immune response in young but not in old men. At baseline, immune response-related genes were higher expressed in old compared to young men. Upstream regulator analyses revealed that most potential regulators were controlling the immune response. Conclusions Based on the gene expression data, we theorise that a short period of CR is not effective in old men regarding immune-related pathways while it is effective in young men. Trial registration ClinicalTrials.gov, NCT00561145 Electronic supplementary material The online version of this article (doi:10.1186/s12263-016-0528-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- I P G Van Bussel
- Division of Human Nutrition, Wageningen University, Bomenweg 2, 6703 HD Wageningen, The Netherlands
| | - A Jolink-Stoppelenburg
- Division of Human Nutrition, Wageningen University, Bomenweg 2, 6703 HD Wageningen, The Netherlands
| | - C P G M De Groot
- Division of Human Nutrition, Wageningen University, Bomenweg 2, 6703 HD Wageningen, The Netherlands
| | - M R Müller
- Division of Human Nutrition, Wageningen University, Bomenweg 2, 6703 HD Wageningen, The Netherlands ; Current Address: Norwich Medical School, University of East Anglia, Norwich, NR4 7TJ UK
| | - L A Afman
- Division of Human Nutrition, Wageningen University, Bomenweg 2, 6703 HD Wageningen, The Netherlands ; Division of Human Nutrition, Wageningen University & Research centre, PO BOX 8129, NL-6700 EV Wageningen, The Netherlands
| |
Collapse
|
41
|
Calvert S, Tacutu R, Sharifi S, Teixeira R, Ghosh P, de Magalhães JP. A network pharmacology approach reveals new candidate caloric restriction mimetics in C. elegans. Aging Cell 2016; 15:256-66. [PMID: 26676933 PMCID: PMC4783339 DOI: 10.1111/acel.12432] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2015] [Indexed: 01/04/2023] Open
Abstract
Caloric restriction (CR), a reduction in calorie intake without malnutrition, retards aging in several animal models from worms to mammals. Developing CR mimetics, compounds that reproduce the longevity benefits of CR without its side effects, is of widespread interest. Here, we employed the Connectivity Map to identify drugs with overlapping gene expression profiles with CR. Eleven statistically significant compounds were predicted as CR mimetics using this bioinformatics approach. We then tested rapamycin, allantoin, trichostatin A, LY‐294002 and geldanamycin in Caenorhabditis elegans. An increase in lifespan and healthspan was observed for all drugs except geldanamycin when fed to wild‐type worms, but no lifespan effects were observed in eat‐2 mutant worms, a genetic model of CR, suggesting that life‐extending effects may be acting via CR‐related mechanisms. We also treated daf‐16 worms with rapamycin, allantoin or trichostatin A, and a lifespan extension was observed, suggesting that these drugs act via DAF‐16‐independent mechanisms, as would be expected from CR mimetics. Supporting this idea, an analysis of predictive targets of the drugs extending lifespan indicates various genes within CR and longevity networks. We also assessed the transcriptional profile of worms treated with either rapamycin or allantoin and found that both drugs use several specific pathways that do not overlap, indicating different modes of action for each compound. The current work validates the capabilities of this bioinformatic drug repositioning method in the context of longevity and reveals new putative CR mimetics that warrant further studies.
Collapse
Affiliation(s)
- Shaun Calvert
- Integrative Genomics of Ageing Group; Institute of Integrative Biology; University of Liverpool; Liverpool UK
| | - Robi Tacutu
- Integrative Genomics of Ageing Group; Institute of Integrative Biology; University of Liverpool; Liverpool UK
| | - Samim Sharifi
- Integrative Genomics of Ageing Group; Institute of Integrative Biology; University of Liverpool; Liverpool UK
| | - Rute Teixeira
- Integrative Genomics of Ageing Group; Institute of Integrative Biology; University of Liverpool; Liverpool UK
| | - Pratul Ghosh
- Integrative Genomics of Ageing Group; Institute of Integrative Biology; University of Liverpool; Liverpool UK
| | - João Pedro de Magalhães
- Integrative Genomics of Ageing Group; Institute of Integrative Biology; University of Liverpool; Liverpool UK
| |
Collapse
|
42
|
Comparative Meta-Analysis of Transcriptomics Data during Cellular Senescence and In Vivo Tissue Ageing. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:732914. [PMID: 25977747 PMCID: PMC4419258 DOI: 10.1155/2015/732914] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Revised: 03/22/2015] [Accepted: 03/23/2015] [Indexed: 02/06/2023]
Abstract
Several studies have employed DNA microarrays to identify gene expression signatures that mark human ageing; yet the features underlying this complicated phenomenon remain elusive. We thus conducted a bioinformatics meta-analysis on transcriptomics data from human cell- and biopsy-based microarrays experiments studying cellular senescence or in vivo tissue ageing, respectively. We report that coregulated genes in the postmitotic muscle and nervous tissues are classified into pathways involved in cancer, focal adhesion, actin cytoskeleton, MAPK signalling, and metabolism regulation. Genes that are differentially regulated during cellular senescence refer to pathways involved in neurodegeneration, focal adhesion, actin cytoskeleton, proteasome, cell cycle, DNA replication, and oxidative phosphorylation. Finally, we revealed genes and pathways (referring to cancer, Huntington's disease, MAPK signalling, focal adhesion, actin cytoskeleton, oxidative phosphorylation, and metabolic signalling) that are coregulated during cellular senescence and in vivo tissue ageing. The molecular commonalities between cellular senescence and tissue ageing are also highlighted by the fact that pathways that were overrepresented exclusively in the biopsy- or cell-based datasets are modules either of the same reference pathway (e.g., metabolism) or of closely interrelated pathways (e.g., thyroid cancer and melanoma). Our reported meta-analysis has revealed novel age-related genes, setting thus the basis for more detailed future functional studies.
Collapse
|
43
|
Barger JL, Anderson RM, Newton MA, da Silva C, Vann JA, Pugh TD, Someya S, Prolla TA, Weindruch R. A conserved transcriptional signature of delayed aging and reduced disease vulnerability is partially mediated by SIRT3. PLoS One 2015; 10:e0120738. [PMID: 25830335 PMCID: PMC4382298 DOI: 10.1371/journal.pone.0120738] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 02/06/2015] [Indexed: 12/25/2022] Open
Abstract
Aging is the most significant risk factor for a range of diseases, including many cancers, neurodegeneration, cardiovascular disease, and diabetes. Caloric restriction (CR) without malnutrition delays aging in diverse species, and therefore offers unique insights into age-related disease vulnerability. Previous studies suggest that there are shared mechanisms of disease resistance associated with delayed aging, however quantitative support is lacking. We therefore sought to identify a common response to CR in diverse tissues and species and determine whether this signature would reflect health status independent of aging. We analyzed gene expression datasets from eight tissues of mice subjected to CR and identified a common transcriptional signature that includes functional categories of mitochondrial energy metabolism, inflammation and ribosomal structure. This signature is detected in flies, rats, and rhesus monkeys on CR, indicating aspects of CR that are evolutionarily conserved. Detection of the signature in mouse genetic models of slowed aging indicates that it is not unique to CR but rather a common aspect of extended longevity. Mice lacking the NAD-dependent deacetylase SIRT3 fail to induce mitochondrial and anti-inflammatory elements of the signature in response to CR, suggesting a potential mechanism involving SIRT3. The inverse of this transcriptional signature is detected with consumption of a high fat diet, obesity and metabolic disease, and is reversed in response to interventions that decrease disease risk. We propose that this evolutionarily conserved, tissue-independent, transcriptional signature of delayed aging and reduced disease vulnerability is a promising target for developing therapies for age-related diseases.
Collapse
Affiliation(s)
- Jamie L. Barger
- LifeGen Technologies LLC, Madison, Wisconsin, United States of America
- * E-mail:
| | - Rozalyn M. Anderson
- Department of Medicine, SMPH, University of Wisconsin, Madison, Wisconsin, United States of America
- Geriatric Research, Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States of America
| | - Michael A. Newton
- Departments of Statistics and of Biostatistics and Medical Informatics, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Cristina da Silva
- LifeGen Technologies LLC, Madison, Wisconsin, United States of America
| | - James A. Vann
- Departments of Genetics and Medical Genetics, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Thomas D. Pugh
- Department of Medicine, SMPH, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Shinichi Someya
- Departments of Genetics and Medical Genetics, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Tomas A. Prolla
- LifeGen Technologies LLC, Madison, Wisconsin, United States of America
- Departments of Genetics and Medical Genetics, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Richard Weindruch
- LifeGen Technologies LLC, Madison, Wisconsin, United States of America
- Department of Medicine, SMPH, University of Wisconsin, Madison, Wisconsin, United States of America
- Geriatric Research, Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States of America
| |
Collapse
|
44
|
Wan C, Freitas AA, de Magalhães JP. Predicting the Pro-Longevity or Anti-Longevity Effect of Model Organism Genes with New Hierarchical Feature Selection Methods. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2015; 12:262-275. [PMID: 26357215 DOI: 10.1109/tcbb.2014.2355218] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Ageing is a highly complex biological process that is still poorly understood. With the growing amount of ageing-related data available on the web, in particular concerning the genetics of ageing, it is timely to apply data mining methods to that data, in order to try to discover novel patterns that may assist ageing research. In this work, we introduce new hierarchical feature selection methods for the classification task of data mining and apply them to ageing-related data from four model organisms: Caenorhabditis elegans (worm), Saccharomyces cerevisiae (yeast), Drosophila melanogaster (fly), and Mus musculus (mouse). The main novel aspect of the proposed feature selection methods is that they exploit hierarchical relationships in the set of features (Gene Ontology terms) in order to improve the predictive accuracy of the Naïve Bayes and 1-Nearest Neighbour (1-NN) classifiers, which are used to classify model organisms' genes into pro-longevity or anti-longevity genes. The results show that our hierarchical feature selection methods, when used together with Naïve Bayes and 1-NN classifiers, obtain higher predictive accuracy than the standard (without feature selection) Naïve Bayes and 1-NN classifiers, respectively. We also discuss the biological relevance of a number of Gene Ontology terms very frequently selected by our algorithms in our datasets.
Collapse
|
45
|
Callahan A, Cifuentes JJ, Dumontier M. An evidence-based approach to identify aging-related genes in Caenorhabditis elegans. BMC Bioinformatics 2015; 16:40. [PMID: 25888240 PMCID: PMC4339751 DOI: 10.1186/s12859-015-0469-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 01/15/2015] [Indexed: 12/21/2022] Open
Abstract
Background Extensive studies have been carried out on Caenorhabditis elegans as a model organism to elucidate mechanisms of aging and the effects of perturbing known aging-related genes on lifespan and behavior. This research has generated large amounts of experimental data that is increasingly difficult to integrate and analyze with existing databases and domain knowledge. To address this challenge, we demonstrate a scalable and effective approach for automatic evidence gathering and evaluation that leverages existing experimental data and literature-curated facts to identify genes involved in aging and lifespan regulation in C. elegans. Results We developed a semantic knowledge base for aging by integrating data about C. elegans genes from WormBase with data about 2005 human and model organism genes from GenAge and 149 genes from GenDR, and with the Bio2RDF network of linked data for the life sciences. Using HyQue (a Semantic Web tool for hypothesis-based querying and evaluation) to interrogate this knowledge base, we examined 48,231 C. elegans genes for their role in modulating lifespan and aging. HyQue identified 24 novel but well-supported candidate aging-related genes for further experimental validation. Conclusions We use semantic technologies to discover candidate aging genes whose effects on lifespan are not yet well understood. Our customized HyQue system, the aging research knowledge base it operates over, and HyQue evaluations of all C. elegans genes are freely available at http://hyque.semanticscience.org. Electronic supplementary material The online version of this article (doi:10.1186/s12859-015-0469-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alison Callahan
- Stanford Center for Biomedical Informatics Research, School of Medicine, Stanford University, Stanford California, AC, USA.
| | - Juan José Cifuentes
- Molecular Bioinformatics Laboratory, Millennium Institute on Immunology and Immunotherapy, 49 Santiago, CP, 8330025, Portugal. .,Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, Santiago, Chile.
| | - Michel Dumontier
- Stanford Center for Biomedical Informatics Research, School of Medicine, Stanford University, Stanford California, AC, USA.
| |
Collapse
|
46
|
Abstract
Dietary restriction (DR) reduces age-specific mortality and increases lifespan in many organisms. DR elicits a large number of physiological changes, however many are undoubtedly not related to longevity. Whole-genome gene expression studies have typically revealed hundreds to thousands of differentially expressed genes in response to DR, and a key open question is which subset of genes mediates longevity. Here we performed transcriptional profiling of fruit flies in a closely spaced time series immediately following a switch to the DR regime and identified four patterns of transcriptional dynamics. Most informatively we find 144 genes rapidly switched to the same level observed in the DR cohort and are hence strong candidates as proximal mediators of reduced mortality upon DR. This class was enriched for genes involved in carbohydrate and fatty acid metabolism. Folate biosynthesis was the only pathway enriched for gene up-regulated upon DR. Four among the down-regulated genes are involved in key regulatory steps within the pentose phosphate pathway, which has been previously associated with lifespan extension in Drosophila. Combined analysis of dietary switch with whole-genome time-course profiling can identify transcriptional responses that are closely associated with and perhaps causal to longevity assurance conferred by dietary restriction.
Collapse
|
47
|
Brenner WG, Schmülling T. Summarizing and exploring data of a decade of cytokinin-related transcriptomics. FRONTIERS IN PLANT SCIENCE 2015; 6:29. [PMID: 25741346 PMCID: PMC4330702 DOI: 10.3389/fpls.2015.00029] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 01/13/2015] [Indexed: 05/17/2023]
Abstract
The genome-wide transcriptional response of the model organism Arabidopsis thaliana to cytokinin has been investigated by different research groups as soon as large-scale transcriptomic techniques became affordable. Over the last 10 years many transcriptomic datasets related to cytokinin have been generated using different technological platforms, some of which are published only in databases, culminating in an RNA sequencing experiment. Two approaches have been made to establish a core set of cytokinin-regulated transcripts by meta-analysis of these datasets using different preferences regarding their selection. Here we add another meta-analysis derived from an independent microarray platform (CATMA), combine all the meta-analyses available with RNAseq data in order to establish an advanced core set of cytokinin-regulated transcripts, and compare the results with the regulation of orthologous rice genes by cytokinin. We discuss the functions of some of the less known cytokinin-regulated genes indicating areas deserving further research to explore cytokinin function. Finally, we investigate the promoters of the core set of cytokinin-induced genes for the abundance and distribution of known cytokinin-responsive cis elements and identify a set of novel candidate motifs.
Collapse
Affiliation(s)
- Wolfram G. Brenner
- *Correspondence: Wolfram G. Brenner and Thomas Schmülling, Dahlem Centre of Plant Sciences, Institute of Biology/Applied Genetics, Freie Universität Berlin, Albrecht-Thaer-Weg 6, D-14195 Berlin, Germany e-mail: ;
| | - Thomas Schmülling
- *Correspondence: Wolfram G. Brenner and Thomas Schmülling, Dahlem Centre of Plant Sciences, Institute of Biology/Applied Genetics, Freie Universität Berlin, Albrecht-Thaer-Weg 6, D-14195 Berlin, Germany e-mail: ;
| |
Collapse
|
48
|
Gebre S, Connor R, Xia Y, Jawed S, Bush JM, Bard M, Elsalloukh H, Tang F. Osh6 overexpression extends the lifespan of yeast by increasing vacuole fusion. Cell Cycle 2014; 11:2176-88. [DOI: 10.4161/cc.20691] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
49
|
Salminen A, Kauppinen A, Hiltunen M, Kaarniranta K. Krebs cycle intermediates regulate DNA and histone methylation: epigenetic impact on the aging process. Ageing Res Rev 2014; 16:45-65. [PMID: 24910305 DOI: 10.1016/j.arr.2014.05.004] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 05/20/2014] [Accepted: 05/29/2014] [Indexed: 02/01/2023]
Abstract
Many aging theories have proposed that mitochondria and energy metabolism have a major role in the aging process. There are recent studies indicating that Krebs cycle intermediates can shape the epigenetic landscape of chromatin by regulating DNA and histone methylation. A growing evidence indicates that epigenetics plays an important role in the regulation of healthspan but also is involved in the aging process. 2-Oxoglutarate (α-ketoglutarate) is a key metabolite in the Krebs cycle but it is also an obligatory substrate for 2-oxoglutarate-dependent dioxygenases (2-OGDO). The 2-OGDO enzyme family includes the major enzymes of DNA and histone demethylation, i.e. Ten-Eleven Translocation (TETs) and Jumonji C domain containing (JmjC) demethylases. In addition, 2-OGDO members can regulate collagen synthesis and hypoxic responses in a non-epigenetical manner. Interestingly, succinate and fumarate, also Krebs cycle intermediates, are potent inhibitors of 2-OGDO enzymes, i.e. the balance of Krebs cycle reactions can affect the level of DNA and histone methylation and thus control gene expression. We will review the epigenetic mechanisms through which Krebs cycle intermediates control the DNA and histone methylation. We propose that age-related disturbances in the Krebs cycle function induce stochastic epigenetic changes in chromatin structures which in turn promote the aging process.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FIN-70211 Kuopio, Finland; Department of Neurology, Kuopio University Hospital, P.O. Box 1777, FIN-70211 Kuopio, Finland.
| | - Anu Kauppinen
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FIN-70211 Kuopio, Finland; Department of Ophthalmology, Kuopio University Hospital, P.O. Box 1777, FIN-70211 Kuopio, Finland
| | - Mikko Hiltunen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FIN-70211 Kuopio, Finland; Department of Neurology, Kuopio University Hospital, P.O. Box 1777, FIN-70211 Kuopio, Finland
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FIN-70211 Kuopio, Finland; Department of Ophthalmology, Kuopio University Hospital, P.O. Box 1777, FIN-70211 Kuopio, Finland
| |
Collapse
|
50
|
Avanesov AS, Ma S, Pierce KA, Yim SH, Lee BC, Clish CB, Gladyshev VN. Age- and diet-associated metabolome remodeling characterizes the aging process driven by damage accumulation. eLife 2014; 3:e02077. [PMID: 24843015 PMCID: PMC4003482 DOI: 10.7554/elife.02077] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Aging is thought to be associated with increased molecular damage, but representative markers vary across conditions and organisms, making it difficult to assess properties of cumulative damage throughout lifespan. We used nontargeted metabolite profiling to follow age-associated trajectories of >15,000 metabolites in Drosophila subjected to control and lifespan-extending diets. We find that aging is associated with increased metabolite diversity and low-abundance molecules, suggesting they include cumulative damage. Remarkably, the number of detected compounds leveled-off in late-life, and this pattern associated with survivorship. Fourteen percent of metabolites showed age-associated changes, which decelerated in late-life and long-lived flies. In contrast, known metabolites changed in abundance similarly to nontargeted metabolites and transcripts, but did not increase in diversity. Targeted profiling also revealed slower metabolism and accumulation of lifespan-limiting molecules. Thus, aging is characterized by gradual metabolome remodeling, and condition- and advanced age-associated deceleration of this remodeling is linked to mortality and molecular damage.DOI: http://dx.doi.org/10.7554/eLife.02077.001.
Collapse
Affiliation(s)
- Andrei S Avanesov
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Siming Ma
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | | | - Sun Hee Yim
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Byung Cheon Lee
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | | | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, United States Broad Institute, Cambridge, United States
| |
Collapse
|