51
|
Giuliani C, Sazzini M, Pirazzini C, Bacalini MG, Marasco E, Ruscone GAG, Fang F, Sarno S, Gentilini D, Di Blasio AM, Crocco P, Passarino G, Mari D, Monti D, Nacmias B, Sorbi S, Salvarani C, Catanoso M, Pettener D, Luiselli D, Ukraintseva S, Yashin A, Franceschi C, Garagnani P. Impact of demography and population dynamics on the genetic architecture of human longevity. Aging (Albany NY) 2019; 10:1947-1963. [PMID: 30089705 PMCID: PMC6128422 DOI: 10.18632/aging.101515] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 07/26/2018] [Indexed: 02/07/2023]
Abstract
The study of the genetics of longevity has been mainly addressed by GWASs that considered subjects from different populations to reach higher statistical power. The "price to pay" is that population-specific evolutionary histories and trade-offs were neglected in the investigation of gene-environment interactions. We propose a new “diachronic” approach that considers processes occurred at both evolutionary and lifespan timescales. We focused on a well-characterized population in terms of evolutionary history (i.e. Italians) and we generated genome-wide data for 333 centenarians from the peninsula and 773 geographically-matched healthy individuals. Obtained results showed that: (i) centenarian genomes are enriched for an ancestral component likely shaped by pre-Neolithic migrations; (ii) centenarians born in Northern Italy unexpectedly clustered with controls from Central/Southern Italy suggesting that Neolithic and Bronze Age gene flow did not favor longevity in this population; (iii) local past adaptive events in response to pathogens and targeting arachidonic acid metabolism became favorable for longevity; (iv) lifelong changes in the frequency of several alleles revealed pleiotropy and trade-off mechanisms crucial for longevity. Therefore, we propose that demographic history and ancient/recent population dynamics need to be properly considered to identify genes involved in longevity, which can differ in different temporal/spatial settings.
Collapse
Affiliation(s)
- Cristina Giuliani
- Department of Biological, Geological, and Environmental Sciences (BiGeA), Laboratory of Molecular Anthropology and Centre for Genome Biology, University of Bologna, Bologna, Italy.,School of Anthropology and Museum Ethnography, University of Oxford, Oxford, UK.,Interdepartmental Center "L. Galvani," (CIG), University of Bologna, Bologna, Italy
| | - Marco Sazzini
- Department of Biological, Geological, and Environmental Sciences (BiGeA), Laboratory of Molecular Anthropology and Centre for Genome Biology, University of Bologna, Bologna, Italy
| | - Chiara Pirazzini
- IRCCS, Institute of Neurological Sciences of Bologna, Bologna, Italy
| | | | - Elena Marasco
- Interdepartmental Center "L. Galvani," (CIG), University of Bologna, Bologna, Italy.,Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy.,Applied Biomedical Research Center (CRBA), S. Orsola-Malpighi Polyclinic, Bologna, Italy
| | - Guido Alberto Gnecchi Ruscone
- Department of Biological, Geological, and Environmental Sciences (BiGeA), Laboratory of Molecular Anthropology and Centre for Genome Biology, University of Bologna, Bologna, Italy
| | - Fang Fang
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC 27708, USA
| | - Stefania Sarno
- Department of Biological, Geological, and Environmental Sciences (BiGeA), Laboratory of Molecular Anthropology and Centre for Genome Biology, University of Bologna, Bologna, Italy
| | - Davide Gentilini
- Istituto Auxologico Italiano IRCCS, Cusano Milanino, Milan, Italy
| | | | - Paolina Crocco
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Giuseppe Passarino
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Daniela Mari
- Geriatric Unit, Department of Medical Sciences and Community Health, Milan, Italy.,Fondazione Ca' Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Daniela Monti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Sandro Sorbi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy.,IRCCS Don Gnocchi, Florence, Italy
| | - Carlo Salvarani
- Azienda Ospedaliera-IRCCS, Reggio Emilia, Italy.,Department of Surgical, Medical, Dental and Morphological Sciences with Interest Transplant, Oncological and Regenerative Medicine, , Italy
| | | | - Davide Pettener
- Department of Biological, Geological, and Environmental Sciences (BiGeA), Laboratory of Molecular Anthropology and Centre for Genome Biology, University of Bologna, Bologna, Italy
| | - Donata Luiselli
- Department for the Cultural Heritage (DBC), University of Bologna, Ravenna, Italy
| | - Svetlana Ukraintseva
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC 27708, USA
| | - Anatoliy Yashin
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC 27708, USA
| | - Claudio Franceschi
- IRCCS, Institute of Neurological Sciences of Bologna, Bologna, Italy.,Co-senior authors
| | - Paolo Garagnani
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy.,Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet at Huddinge University Hospital, S-141 86 Stockholm, Sweden.,CNR Institute of Molecular Genetics, Unit of Bologna, Bologna, Italy.,Rizzoli Orthopaedic Institute, Laboratory of Cell Biology, Bologna, Italy.,Co-senior authors
| |
Collapse
|
52
|
Giuliani C, Garagnani P, Franceschi C. Genetics of Human Longevity Within an Eco-Evolutionary Nature-Nurture Framework. Circ Res 2019; 123:745-772. [PMID: 30355083 DOI: 10.1161/circresaha.118.312562] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Human longevity is a complex trait, and to disentangle its basis has a great theoretical and practical consequences for biomedicine. The genetics of human longevity is still poorly understood despite several investigations that used different strategies and protocols. Here, we argue that such rather disappointing harvest is largely because of the extraordinary complexity of the longevity phenotype in humans. The capability to reach the extreme decades of human lifespan seems to be the result of an intriguing mixture of gene-environment interactions. Accordingly, the genetics of human longevity is here described as a highly context-dependent phenomenon, within a new integrated, ecological, and evolutionary perspective, and is presented as a dynamic process, both historically and individually. The available literature has been scrutinized within this perspective, paying particular attention to factors (sex, individual biography, family, population ancestry, social structure, economic status, and education, among others) that have been relatively neglected. The strength and limitations of the most powerful and used tools, such as genome-wide association study and whole-genome sequencing, have been discussed, focusing on prominently emerged genes and regions, such as apolipoprotein E, Forkhead box O3, interleukin 6, insulin-like growth factor-1, chromosome 9p21, 5q33.3, and somatic mutations among others. The major results of this approach suggest that (1) the genetics of longevity is highly population specific; (2) small-effect alleles, pleiotropy, and the complex allele timing likely play a major role; (3) genetic risk factors are age specific and need to be integrated in the light of the geroscience perspective; (4) a close relationship between genetics of longevity and genetics of age-related diseases (especially cardiovascular diseases) do exist. Finally, the urgent need of a global approach to the largely unexplored interactions between the 3 genetics of human body, that is, nuclear, mitochondrial, and microbiomes, is stressed. We surmise that the comprehensive approach here presented will help in increasing the above-mentioned harvest.
Collapse
Affiliation(s)
- Cristina Giuliani
- From the Department of Biological, Geological, and Environmental Sciences (BiGeA), Laboratory of Molecular Anthropology and Centre for Genome Biology (C.G.), University of Bologna, Italy.,School of Anthropology and Museum Ethnography, University of Oxford, United Kingdom (C.G.).,Interdepartmental Centre 'L. Galvani' (CIG), University of Bologna, Italy (C.G.)
| | - Paolo Garagnani
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES) (P.G.), University of Bologna, Italy.,Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet at Huddinge University Hospital, Stockholm, Sweden (P.G.)
| | | |
Collapse
|
53
|
Abstract
Cardiac ageing manifests as a decline in function leading to heart failure. At the cellular level, ageing entails decreased replicative capacity and dysregulation of cellular processes in myocardial and nonmyocyte cells. Various extrinsic parameters, such as lifestyle and environment, integrate important signalling pathways, such as those involving inflammation and oxidative stress, with intrinsic molecular mechanisms underlying resistance versus progression to cellular senescence. Mitigation of cardiac functional decline in an ageing organism requires the activation of enhanced maintenance and reparative capacity, thereby overcoming inherent endogenous limitations to retaining a youthful phenotype. Deciphering the molecular mechanisms underlying dysregulation of cellular function and renewal reveals potential interventional targets to attenuate degenerative processes at the cellular and systemic levels to improve quality of life for our ageing population. In this Review, we discuss the roles of extrinsic and intrinsic factors in cardiac ageing. Animal models of cardiac ageing are summarized, followed by an overview of the current and possible future treatments to mitigate the deleterious effects of cardiac ageing.
Collapse
|
54
|
Nygaard HB, Erson-Omay EZ, Wu X, Kent BA, Bernales CQ, Evans DM, Farrer MJ, Vilariño-Güell C, Strittmatter SM. Whole-Exome Sequencing of an Exceptional Longevity Cohort. J Gerontol A Biol Sci Med Sci 2019; 74:1386-1390. [PMID: 29750252 PMCID: PMC6696723 DOI: 10.1093/gerona/gly098] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/26/2018] [Indexed: 12/22/2022] Open
Abstract
Centenarians represent a unique cohort to study the genetic basis for longevity and factors determining the risk of neurodegenerative disorders, including Alzheimer's disease (AD). The estimated genetic contribution to longevity is highest in centenarians and super-cententenarians, but few genetic variants have been shown to clearly impact this phenotype. While the genetic risk for AD and other dementias is now well understood, the frequency of known dementia risk variants in centenarians is not fully characterized. To address these questions, we performed whole-exome sequencing on 100 individuals of 98-108 years age in search of genes with large effect sizes towards the exceptional aging phenotype. Overall, we were unable to identify a rare protein-altering variant or individual genes with an increased burden of rare variants associated with exceptional longevity. Gene burden analysis revealed three genes of nominal statistical significance associated with extreme aging, including LYST, MDN1, and RBMXL1. Several genes with variants conferring an increased risk for AD and other dementias were identified, including TREM2, EPHA1, ABCA7, PLD3, MAPT, and NOTCH3. Larger centenarian studies will be required to further elucidate the genetic basis for longevity, and factors conferring protection against age-dependent neurodegenerative syndromes.
Collapse
Affiliation(s)
- Haakon B Nygaard
- Division of Neurology, University of British Columbia, Vancouver, British Columbia, Canada
| | - E Zeynep Erson-Omay
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut
| | - Xiujuan Wu
- Division of Neurology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Brianne A Kent
- Division of Neurology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Cecily Q Bernales
- Department of Medical Genetics, Centre for Applied Neurogenetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Daniel M Evans
- Department of Medical Genetics, Centre for Applied Neurogenetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Matthew J Farrer
- Department of Medical Genetics, Centre for Applied Neurogenetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Carles Vilariño-Güell
- Department of Medical Genetics, Centre for Applied Neurogenetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Stephen M Strittmatter
- Program in Cellular Neuroscience, Neurodegeneration and Repair (CNNR), Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
55
|
Gardiner SL, Trompet S, Sabayan B, Boogaard MW, Jukema JW, Slagboom PE, Roos RAC, van der Grond J, Aziz NA. Repeat variations in polyglutamine disease-associated genes and cognitive function in old age. Neurobiol Aging 2019; 84:236.e17-236.e28. [PMID: 31522753 DOI: 10.1016/j.neurobiolaging.2019.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 08/03/2019] [Accepted: 08/04/2019] [Indexed: 02/03/2023]
Abstract
Although the heritability of cognitive function in old age is substantial, genome-wide association studies have had limited success in elucidating its genetic basis, leaving a considerable amount of "missing heritability." Aside from single nucleotide polymorphisms, genome-wide association studies are unable to assess other large sources of genetic variation, such as tandem repeat polymorphisms. Therefore, here, we studied the association of cytosine-adenine-guanine (CAG) repeat variations in polyglutamine disease-associated genes (PDAGs) with cognitive function in older adults. In a large cohort consisting of 5786 participants, we found that the CAG repeat number in 3 PDAGs (TBP, HTT, and AR) were significantly associated with the decline in cognitive function, which together accounted for 0.49% of the variation. Furthermore, in an magnetic resonance imaging substudy, we found that CAG repeat polymorphisms in 4 PDAGs (ATXN2, CACNA1A, ATXN7, and AR) were associated with different imaging characteristics, including brain stem, putamen, globus pallidus, thalamus, and amygdala volumes. Our findings indicate that tandem repeat polymorphisms are associated with cognitive function in older adults and highlight the importance of PDAGs in elucidating its missing heritability.
Collapse
Affiliation(s)
- Sarah L Gardiner
- Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands; Department of Human Genetics, Leiden University Medical Centre, Leiden, the Netherlands.
| | - Stella Trompet
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Centre, Leiden, the Netherlands
| | - Behnam Sabayan
- The Ken and Ruth Davee Department of Neurology, Northwestern University, Chicago, IL, USA
| | - Merel W Boogaard
- Department of Clinical Genetics, Leiden University Medical Centre, Leiden, the Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Centre, Leiden, the Netherlands
| | - P Eline Slagboom
- Department of Molecular Epidemiology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Raymund A C Roos
- Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Jeroen van der Grond
- Department of Radiology, Leiden University Medical Centre, Leiden, the Netherlands
| | - N Ahmad Aziz
- Population Health Sciences, German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany; Department of Neurology, University of Bonn, Bonn, Germany
| |
Collapse
|
56
|
Morris BJ, Willcox BJ, Donlon TA. Genetic and epigenetic regulation of human aging and longevity. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1718-1744. [PMID: 31109447 PMCID: PMC7295568 DOI: 10.1016/j.bbadis.2018.08.039] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 08/02/2018] [Accepted: 08/28/2018] [Indexed: 02/06/2023]
Abstract
Here we summarize the latest data on genetic and epigenetic contributions to human aging and longevity. Whereas environmental and lifestyle factors are important at younger ages, the contribution of genetics appears more important in reaching extreme old age. Genome-wide studies have implicated ~57 gene loci in lifespan. Epigenomic changes during aging profoundly affect cellular function and stress resistance. Dysregulation of transcriptional and chromatin networks is likely a crucial component of aging. Large-scale bioinformatic analyses have revealed involvement of numerous interaction networks. As the young well-differentiated cell replicates into eventual senescence there is drift in the highly regulated chromatin marks towards an entropic middle-ground between repressed and active, such that genes that were previously inactive "leak". There is a breakdown in chromatin connectivity such that topologically associated domains and their insulators weaken, and well-defined blocks of constitutive heterochromatin give way to generalized, senescence-associated heterochromatin, foci. Together, these phenomena contribute to aging.
Collapse
Affiliation(s)
- Brian J Morris
- Basic & Clinical Genomics Laboratory, School of Medical Sciences and Bosch Institute, University of Sydney, New South Wales 2006, Australia; Honolulu Heart Program (HHP)/Honolulu-Asia Aging Study (HAAS), Department of Research, Kuakini Medical Center, Honolulu, HI 96817, United States; Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Kuakini Medical Center Campus, Honolulu, HI 96813, United States.
| | - Bradley J Willcox
- Honolulu Heart Program (HHP)/Honolulu-Asia Aging Study (HAAS), Department of Research, Kuakini Medical Center, Honolulu, HI 96817, United States; Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Kuakini Medical Center Campus, Honolulu, HI 96813, United States.
| | - Timothy A Donlon
- Honolulu Heart Program (HHP)/Honolulu-Asia Aging Study (HAAS), Department of Research, Kuakini Medical Center, Honolulu, HI 96817, United States; Departments of Cell & Molecular Biology and Pathology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, United States.
| |
Collapse
|
57
|
Fan CC, Smeland OB, Schork AJ, Chen CH, Holland D, Lo MT, Sundar VS, Frei O, Jernigan TL, Andreassen OA, Dale AM. Beyond heritability: improving discoverability in imaging genetics. Hum Mol Genet 2019. [PMID: 29522091 DOI: 10.1093/hmg/ddy082] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Structural neuroimaging measures based on magnetic resonance imaging have been at the forefront of imaging genetics. Global efforts to ensure homogeneity of measurements across study sites have enabled large-scale imaging genetic projects, accumulating nearly 50K samples for genome-wide association studies (GWAS). However, not many novel genetic variants have been identified by these GWAS, despite the high heritability of structural neuroimaging measures. Here, we discuss the limitations of using heritability as a guidance for assessing statistical power of GWAS, and highlight the importance of discoverability-which is the power to detect genetic variants for a given phenotype depending on its unique genomic architecture and GWAS sample size. Further, we present newly developed methods that boost genetic discovery in imaging genetics. By redefining imaging measures independent of traditional anatomical conventions, it is possible to improve discoverability, enabling identification of more genetic effects. Moreover, by leveraging enrichment priors from genomic annotations and independent GWAS of pleiotropic traits, we can better characterize effect size distributions, and identify reliable and replicable loci associated with structural neuroimaging measures. Statistical tools leveraging novel insights into the genetic discoverability of human traits, promises to accelerate the identification of genetic underpinnings underlying brain structural variation.
Collapse
Affiliation(s)
- Chun Chieh Fan
- Center for Multimodal Imaging and Genetics, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Olav B Smeland
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Andrew J Schork
- Institute for Biological Psychiatry, Mental Health Center Sct. Hans, Capital Region of Denmark, Denmark
| | - Chi-Hua Chen
- Center for Multimodal Imaging and Genetics, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA.,Department of Radiology, School of Medicine, University of California San Diego, La Jolla, CA 92037, USA
| | - Dominic Holland
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA 92037, USA
| | - Min-Tzu Lo
- Center for Multimodal Imaging and Genetics, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA.,Department of Radiology, School of Medicine, University of California San Diego, La Jolla, CA 92037, USA
| | - V S Sundar
- Center for Multimodal Imaging and Genetics, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA.,Department of Radiology, School of Medicine, University of California San Diego, La Jolla, CA 92037, USA
| | - Oleksandr Frei
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Terry L Jernigan
- Center for Human Development, University of California San Diego, La Jolla, CA 92093, USA
| | - Ole A Andreassen
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Anders M Dale
- Center for Multimodal Imaging and Genetics, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA.,Department of Radiology, School of Medicine, University of California San Diego, La Jolla, CA 92037, USA.,Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA 92037, USA
| |
Collapse
|
58
|
Kulminski AM, Loika Y, Huang J, Arbeev KG, Bagley O, Ukraintseva S, Yashin AI, Culminskaya I. Pleiotropic Meta-Analysis of Age-Related Phenotypes Addressing Evolutionary Uncertainty in Their Molecular Mechanisms. Front Genet 2019; 10:433. [PMID: 31134135 PMCID: PMC6524409 DOI: 10.3389/fgene.2019.00433] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 04/24/2019] [Indexed: 12/21/2022] Open
Abstract
Age-related phenotypes are characterized by genetic heterogeneity attributed to an uncertain role of evolution in establishing their molecular mechanisms. Here, we performed univariate and pleiotropic meta-analyses of 24 age-related phenotypes dealing with such evolutionary uncertainty and leveraging longitudinal information. Our analysis identified 237 novel single nucleotide polymorphisms (SNPs) in 199 loci with phenotype-specific (61 SNPs) and pleiotropic (176 SNPs) associations and replicated associations for 160 SNPs in 68 loci in a modest sample of 26,371 individuals from five longitudinal studies. Most pleiotropic associations (65.3%, 115 of 176 SNPs) were impacted by heterogeneity, with the natural-selection—free genetic heterogeneity as its inevitable component. This pleiotropic heterogeneity was dominated (93%, 107 of 115 SNPs) by antagonistic genetic heterogeneity, a phenomenon that is characterized by antagonistic directions of genetic effects for directly correlated phenotypes. Genetic association studies of age-related phenotypes addressing the evolutionary uncertainty in establishing their molecular mechanisms have power to substantially improve the efficiency of the analyses. A dominant form of heterogeneous pleiotropy, antagonistic genetic heterogeneity, provides unprecedented insight into the genetic origin of age-related phenotypes and side effects in medical care that is counter-intuitive in medical genetics but naturally expected when molecular mechanisms of age-related phenotypes are not due to direct evolutionary selection.
Collapse
Affiliation(s)
- Alexander M Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Yury Loika
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Jian Huang
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Konstantin G Arbeev
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Olivia Bagley
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Svetlana Ukraintseva
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Anatoliy I Yashin
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Irina Culminskaya
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| |
Collapse
|
59
|
Singh PP, Demmitt BA, Nath RD, Brunet A. The Genetics of Aging: A Vertebrate Perspective. Cell 2019; 177:200-220. [PMID: 30901541 PMCID: PMC7592626 DOI: 10.1016/j.cell.2019.02.038] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 02/21/2019] [Accepted: 02/22/2019] [Indexed: 02/07/2023]
Abstract
Aging negatively impacts vitality and health. Many genetic pathways that regulate aging were discovered in invertebrates. However, the genetics of aging is more complex in vertebrates because of their specialized systems. This Review discusses advances in the genetic regulation of aging in vertebrates from work in mice, humans, and organisms with exceptional lifespans. We highlight challenges for the future, including sex-dependent differences in lifespan and the interplay between genes and environment. We also discuss how the identification of reliable biomarkers of age and development of new vertebrate models can be leveraged for personalized interventions to counter aging and age-related diseases.
Collapse
Affiliation(s)
- Param Priya Singh
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | | | - Ravi D Nath
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Anne Brunet
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Glenn Laboratories for the Biology of Aging, Stanford, CA 94305, USA.
| |
Collapse
|
60
|
Tony Cai T, Sun W, Wang W. Covariate‐assisted ranking and screening for large‐scale two‐sample inference. J R Stat Soc Series B Stat Methodol 2019. [DOI: 10.1111/rssb.12304] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
| | - Wenguang Sun
- University of Southern California Los Angeles USA
| | - Weinan Wang
- University of Southern California Los Angeles USA
| |
Collapse
|
61
|
The Genetic Variability of APOE in Different Human Populations and Its Implications for Longevity. Genes (Basel) 2019; 10:genes10030222. [PMID: 30884759 PMCID: PMC6471373 DOI: 10.3390/genes10030222] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/01/2019] [Accepted: 03/12/2019] [Indexed: 12/11/2022] Open
Abstract
Human longevity is a complex phenotype resulting from the combinations of context-dependent gene-environment interactions that require analysis as a dynamic process in a cohesive ecological and evolutionary framework. Genome-wide association (GWAS) and whole-genome sequencing (WGS) studies on centenarians pointed toward the inclusion of the apolipoprotein E (APOE) polymorphisms ε2 and ε4, as implicated in the attainment of extreme longevity, which refers to their effect in age-related Alzheimer's disease (AD) and cardiovascular disease (CVD). In this case, the available literature on APOE and its involvement in longevity is described according to an anthropological and population genetics perspective. This aims to highlight the evolutionary history of this gene, how its participation in several biological pathways relates to human longevity, and which evolutionary dynamics may have shaped the distribution of APOE haplotypes across the globe. Its potential adaptive role will be described along with implications for the study of longevity in different human groups. This review also presents an updated overview of the worldwide distribution of APOE alleles based on modern day data from public databases and ancient DNA samples retrieved from literature in the attempt to understand the spatial and temporal frame in which present-day patterns of APOE variation evolved.
Collapse
|
62
|
Zenin A, Tsepilov Y, Sharapov S, Getmantsev E, Menshikov LI, Fedichev PO, Aulchenko Y. Identification of 12 genetic loci associated with human healthspan. Commun Biol 2019; 2:41. [PMID: 30729179 PMCID: PMC6353874 DOI: 10.1038/s42003-019-0290-0] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 01/08/2019] [Indexed: 02/06/2023] Open
Abstract
Aging populations face diminishing quality of life due to increased disease and morbidity. These challenges call for longevity research to focus on understanding the pathways controlling healthspan. We use the data from the UK Biobank (UKB) cohort and observe that the risks of major chronic diseases increased exponentially and double every eight years, i.e., at a rate compatible with the Gompertz mortality law. Assuming that aging drives the acceleration in morbidity rates, we build a risk model to predict the age at the end of healthspan depending on age, gender, and genetic background. Using the sub-population of 300,447 British individuals as a discovery cohort, we identify 12 loci associated with healthspan at the whole-genome significance level. We find strong genetic correlations between healthspan and all-cause mortality, life-history, and lifestyle traits. We thereby conclude that the healthspan offers a promising new way to interrogate the genetics of human longevity.
Collapse
Affiliation(s)
- Aleksandr Zenin
- Gero LLC, Novokuznetskaya street 24/2, Moscow, Russia 119017
| | - Yakov Tsepilov
- Novosibirsk State University, Pirogova 2, Novosibirsk, Russia 630090
- Institute of Cytology and Genetics SB RAS, Lavrentyeva ave. 10, Novosibirsk, Russia 630090
| | - Sodbo Sharapov
- Novosibirsk State University, Pirogova 2, Novosibirsk, Russia 630090
- Institute of Cytology and Genetics SB RAS, Lavrentyeva ave. 10, Novosibirsk, Russia 630090
| | | | - L. I. Menshikov
- Gero LLC, Novokuznetskaya street 24/2, Moscow, Russia 119017
- National Research Center “Kurchatov Institute”, 1, Akademika Kurchatova pl., Moscow, Russia 123182
| | - Peter O. Fedichev
- Gero LLC, Novokuznetskaya street 24/2, Moscow, Russia 119017
- Moscow Institute of Physics and Technology, Institutskii per. 9, Dolgoprudny, Moscow Russia 141700
| | - Yurii Aulchenko
- Novosibirsk State University, Pirogova 2, Novosibirsk, Russia 630090
- Institute of Cytology and Genetics SB RAS, Lavrentyeva ave. 10, Novosibirsk, Russia 630090
- PolyOmica, Het Vlaggeschip 61, 5237PA ‘s-Hertogenbosch, The Netherlands
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh, Scotland EH8 9AG UK
| |
Collapse
|
63
|
Cells exhibiting strong p16 INK4a promoter activation in vivo display features of senescence. Proc Natl Acad Sci U S A 2019; 116:2603-2611. [PMID: 30683717 PMCID: PMC6377452 DOI: 10.1073/pnas.1818313116] [Citation(s) in RCA: 230] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The accumulation of senescent cells over a lifetime causes age-related pathologies; however, the inability to reliably identify senescent cells in vivo has hindered clinical efforts to employ this knowledge as a means to ameliorate or reverse aging. Here, we describe a reporter allele, p16tdTom, enabling the in vivo identification and isolation of cells featuring high-level activation of the p16INK4a promoter. Our findings provide an insight into the functional and molecular characteristics of p16INK4a-activated cells in vitro and in vivo. We show that such cells accumulate with aging or other models of injury, and that they exhibit clinically targetable features of cellular senescence. The activation of cellular senescence throughout the lifespan promotes tumor suppression, whereas the persistence of senescent cells contributes to aspects of aging. This theory has been limited, however, by an inability to identify and isolate individual senescent cells within an intact organism. Toward that end, we generated a murine reporter strain by “knocking-in” a fluorochrome, tandem-dimer Tomato (tdTom), into exon 1α of the p16INK4a locus. We used this allele (p16tdTom) for the enumeration, isolation, and characterization of individual p16INK4a-expressing cells (tdTom+). The half-life of the knocked-in transcript was shorter than that of the endogenous p16INK4a mRNA, and therefore reporter expression better correlated with p16INK4a promoter activation than p16INK4a transcript abundance. The frequency of tdTom+ cells increased with serial passage in cultured murine embryo fibroblasts from p16tdTom/+ mice. In adult mice, tdTom+ cells could be readily detected at low frequency in many tissues, and the frequency of these cells increased with aging. Using an in vivo model of peritoneal inflammation, we compared the phenotype of cells with or without activation of p16INK4a and found that tdTom+ macrophages exhibited some features of senescence, including reduced proliferation, senescence-associated β-galactosidase (SA-β-gal) activation, and increased mRNA expression of a subset of transcripts encoding factors involved in SA-secretory phenotype (SASP). These results indicate that cells harboring activation of the p16INK4a promoter accumulate with aging and inflammation in vivo, and display characteristics of senescence.
Collapse
|
64
|
Ferri E, Gussago C, Casati M, Mari D, Rossi PD, Ciccone S, Cesari M, Arosio B. Apolipoprotein E gene in physiological and pathological aging. Mech Ageing Dev 2019; 178:41-45. [PMID: 30658061 DOI: 10.1016/j.mad.2019.01.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 01/02/2019] [Accepted: 01/15/2019] [Indexed: 01/03/2023]
Abstract
INTRODUCTION The genetic background plays a role on longevity. The distribution of the apolipoprotein E gene (APOE) variants (ε2, ε3, ε4) may differ across age groups, especially in the oldest old and despite geographical and ethnic specificities. Since the ε4 variant is associated with Alzheimer's disease (AD), it might represent an opportunity for exploring the relationship of APOE with physiological and pathological aging. AIM To explore the role played by APOE genotype/alleles on physiological and pathological brain aging. MATERIALS AND METHODS The study was conducted in a cohort of centenarians (n = 106), and two cohorts of octogenarians (without cognitive decline, n = 351 controls; and with AD, n = 294). RESULTS No significant differences in genotype/allele distributions were observed comparing controls to centenarians. The prevalence of ε2/ε3, ε3/ε3, ε3/ε4 and ε4/ε4 genotypes were significantly different in centenarians compared to AD. The prevalence of ε2 and ε3 alleles were significantly higher in centenarians, whereas the ε4 was less frequent. The ε4 allele was positively associated with AD, whereas a negative association was found for ε2 and ε3 alleles. CONCLUSIONS Our study indicates that ε4 allele is strongly associated with AD. APOE significantly affects AD risk, but apparently not longevity.
Collapse
Affiliation(s)
- E Ferri
- Geriatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Pace 9, 20122 Milan, Italy.
| | - C Gussago
- Department of Clinical Sciences and Community Health, University of Milan, Via Pace 9, 20122 Milan, Italy.
| | - M Casati
- Geriatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Pace 9, 20122 Milan, Italy.
| | - D Mari
- Geriatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Pace 9, 20122 Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, Via Pace 9, 20122 Milan, Italy.
| | - P D Rossi
- Geriatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Pace 9, 20122 Milan, Italy.
| | - S Ciccone
- Geriatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Pace 9, 20122 Milan, Italy.
| | - M Cesari
- Geriatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Pace 9, 20122 Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, Via Pace 9, 20122 Milan, Italy.
| | - B Arosio
- Geriatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Pace 9, 20122 Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, Via Pace 9, 20122 Milan, Italy.
| |
Collapse
|
65
|
Serbezov D, Balabanski L, Hadjidekova S, Toncheva D. Genomics of longevity: recent insights from research on centenarians. BIOTECHNOL BIOTEC EQ 2018. [DOI: 10.1080/13102818.2018.1532317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Affiliation(s)
- Dimitar Serbezov
- Department of Medical Genetics, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Lubomir Balabanski
- Department of Medical Genetics, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
- Assisted Reproduction Clinic “Dr. Malinov”, Sofia, Bulgaria
| | - Savina Hadjidekova
- Department of Medical Genetics, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Draga Toncheva
- Department of Medical Genetics, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
- Assisted Reproduction Clinic “Dr. Malinov”, Sofia, Bulgaria
| |
Collapse
|
66
|
Hook M, Roy S, Williams EG, Bou Sleiman M, Mozhui K, Nelson JF, Lu L, Auwerx J, Williams RW. Genetic cartography of longevity in humans and mice: Current landscape and horizons. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2718-2732. [PMID: 29410319 PMCID: PMC6066442 DOI: 10.1016/j.bbadis.2018.01.026] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/24/2018] [Accepted: 01/28/2018] [Indexed: 12/14/2022]
Abstract
Aging is a complex and highly variable process. Heritability of longevity among humans and other species is low, and this finding has given rise to the idea that it may be futile to search for DNA variants that modulate aging. We argue that the problem in mapping longevity genes is mainly one of low power and the genetic and environmental complexity of aging. In this review we highlight progress made in mapping genes and molecular networks associated with longevity, paying special attention to work in mice and humans. We summarize 40 years of linkage studies using murine cohorts and 15 years of studies in human populations that have exploited candidate gene and genome-wide association methods. A small but growing number of gene variants contribute to known longevity mechanisms, but a much larger set have unknown functions. We outline these and other challenges and suggest some possible solutions, including more intense collaboration between research communities that use model organisms and human cohorts. Once hundreds of gene variants have been linked to differences in longevity in mammals, it will become feasible to systematically explore gene-by-environmental interactions, dissect mechanisms with more assurance, and evaluate the roles of epistasis and epigenetics in aging. A deeper understanding of complex networks-genetic, cellular, physiological, and social-should position us well to improve healthspan.
Collapse
Affiliation(s)
- Michael Hook
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Suheeta Roy
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Evan G Williams
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich CH-8093, Switzerland
| | - Maroun Bou Sleiman
- Interfaculty Institute of Bioengineering, Laboratory of Integrative and Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne CH-1015, Switzerland
| | - Khyobeni Mozhui
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - James F Nelson
- Department of Cellular and Integrative Physiology and Barshop Institute for Longevity and Aging Studies, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Lu Lu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Johan Auwerx
- Interfaculty Institute of Bioengineering, Laboratory of Integrative and Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne CH-1015, Switzerland
| | - Robert W Williams
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
67
|
Kulminski AM, Huang J, Wang J, He L, Loika Y, Culminskaya I. Apolipoprotein E region molecular signatures of Alzheimer's disease. Aging Cell 2018; 17:e12779. [PMID: 29797398 PMCID: PMC6052488 DOI: 10.1111/acel.12779] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2018] [Indexed: 01/01/2023] Open
Abstract
Although the APOE region is the strongest genetic risk factor for Alzheimer's diseases (ADs), its pathogenic role remains poorly understood. Elucidating genetic predisposition to ADs, a subset of age-related diseases characteristic for postreproductive period, is hampered by the undefined role of evolution in establishing molecular mechanisms of such diseases. This uncertainty is inevitable source of natural-selection-free genetic heterogeneity in predisposition to ADs. We performed first large-scale analysis of linkage disequilibrium (LD) structures characterized by 30 polymorphisms from five genes in the APOE 19q13.3 region (BCAM, NECTIN2, TOMM40, APOE, and APOC1) in 2,673 AD-affected and 16,246 unaffected individuals from five cohorts. Consistent with the undefined role of evolution in age-related diseases, we found that these structures, being highly heterogeneous, are significantly different in subjects with and without ADs. The pattern of the difference represents molecular signature of AD comprised of single nucleotide polymorphisms (SNPs) from all five genes in the APOE region. Significant differences in LD in subjects with and without ADs indicate SNPs from different genes likely involved in AD pathogenesis. Significant and highly heterogeneous molecular signatures of ADs provide unprecedented insight into complex polygenetic predisposition to ADs in the APOE region. These findings are more consistent with a complex haplotype than with a single genetic variant origin of ADs in this region.
Collapse
Affiliation(s)
- Alexander M Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina
| | - Jian Huang
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina
| | - Jiayi Wang
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina
| | - Liang He
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina
| | - Yury Loika
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina
| | - Irina Culminskaya
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina
| |
Collapse
|
68
|
Stakišaitis D, Juknevičienė M, Ulys A, Žaliūnienė D, Stanislovaitienė D, Šepetienė R, Slavinska A, Sužiedėlis K, Lesauskaitė V. ABO blood group polymorphism has an impact on prostate, kidney and bladder cancer in association with longevity. Oncol Lett 2018; 16:1321-1331. [PMID: 30061952 PMCID: PMC6063046 DOI: 10.3892/ol.2018.8749] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 04/16/2018] [Indexed: 02/07/2023] Open
Abstract
The aim of the present study was to assess the ABO blood group polymorphism association with prostate, bladder and kidney cancer, and longevity. The following data groups were analyzed: Prostate cancer (n=2,200), bladder cancer (n=1,530), renal cell cancer (n=2,650), oldest-old (n=166) and blood donors (n=994) groups. The data on the ABO blood type frequency and odds ratio in prostate cancer patients revealed a significantly higher blood group B frequency (P<0.05); the pooled men and women, separate men bladder cancer risk was significantly associated with the blood group B (P<0.04); however, no such association was identified in the female patients. The blood group O was observed to have a significantly decreased risk of bladder cancer for females (P<0.05). No significance for the ABO blood group type in the studied kidney cancer patients was identified. A comparison of the oldest-old and blood donor groups revealed that blood group A was significantly more frequent and blood type B was significantly rarer in the oldest-olds (P<0.05). The results of the present study indicated that blood type B was associated with the risk of prostate and bladder cancer, and could be evaluated as a determinant in the negative assocation with longevity. Blood types O and A may be positive factors for increasing the oldest-old age likelihood. The clustering analysis by the ABO type frequency demonstrated that the oldest-olds comprised a separate cluster of the studied groups.
Collapse
Affiliation(s)
- Donatas Stakišaitis
- Laboratory of Molecular Oncology, National Cancer Institute, LT-08660 Vilnius, Lithuania
| | - Milda Juknevičienė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| | - Albertas Ulys
- Oncosurgery Clinics, National Cancer Institute, LT-08660 Vilnius, Lithuania
| | - Dalia Žaliūnienė
- Department of Ophtalmology, Medical Academy, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| | - Daiva Stanislovaitienė
- Department of Ophtalmology, Medical Academy, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| | - Ramunė Šepetienė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| | | | - Kęstutis Sužiedėlis
- Laboratory of Molecular Oncology, National Cancer Institute, LT-08660 Vilnius, Lithuania
| | - Vita Lesauskaitė
- Department of Geriatrics, Medical Academy, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| |
Collapse
|
69
|
Tuttle CS, Maier AB. Towards a biological geriatric assessment. Exp Gerontol 2018; 107:102-107. [DOI: 10.1016/j.exger.2017.09.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 09/24/2017] [Accepted: 09/25/2017] [Indexed: 12/30/2022]
|
70
|
Lei L, Fithian W. AdaPT: an interactive procedure for multiple testing with side information. J R Stat Soc Series B Stat Methodol 2018. [DOI: 10.1111/rssb.12274] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Lihua Lei
- University of California; Berkeley USA
| | | |
Collapse
|
71
|
Dobriban E. Weighted mining of massive collections of [Formula: see text]-values by convex optimization. INFORMATION AND INFERENCE : A JOURNAL OF THE IMA 2018; 7:251-275. [PMID: 29930799 PMCID: PMC5998655 DOI: 10.1093/imaiai/iax013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 10/05/2017] [Indexed: 06/08/2023]
Abstract
Researchers in data-rich disciplines-think of computational genomics and observational cosmology-often wish to mine large bodies of [Formula: see text]-values looking for significant effects, while controlling the false discovery rate or family-wise error rate. Increasingly, researchers also wish to prioritize certain hypotheses, for example, those thought to have larger effect sizes, by upweighting, and to impose constraints on the underlying mining, such as monotonicity along a certain sequence. We introduce Princessp, a principled method for performing weighted multiple testing by constrained convex optimization. Our method elegantly allows one to prioritize certain hypotheses through upweighting and to discount others through downweighting, while constraining the underlying weights involved in the mining process. When the [Formula: see text]-values derive from monotone likelihood ratio families such as the Gaussian means model, the new method allows exact solution of an important optimal weighting problem previously thought to be non-convex and computationally infeasible. Our method scales to massive data set sizes. We illustrate the applications of Princessp on a series of standard genomics data sets and offer comparisons with several previous 'standard' methods. Princessp offers both ease of operation and the ability to scale to extremely large problem sizes. The method is available as open-source software from github.com/dobriban/pvalue_weighting_matlab (accessed 11 October 2017).
Collapse
Affiliation(s)
- Edgar Dobriban
- Department of Statistics, The Wharton School, University of Pennsylania, USA
| |
Collapse
|
72
|
Halaschek-Wiener J, Tindale LC, Collins JA, Leach S, McManus B, Madden K, Meneilly G, Le ND, Connors JM, Brooks-Wilson AR. The Super-Seniors Study: Phenotypic characterization of a healthy 85+ population. PLoS One 2018; 13:e0197578. [PMID: 29795606 PMCID: PMC5967696 DOI: 10.1371/journal.pone.0197578] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 05/06/2018] [Indexed: 11/18/2022] Open
Abstract
Background To understand why some people live to advanced age in good health and others do not, it is important to study not only disease, but also long-term good health. The Super-Seniors Study aims to identify factors associated with healthy aging. Methods 480 healthy oldest-old ‘Super-Seniors’ aged 85 to 105 years and never diagnosed with cancer, cardiovascular disease, diabetes, dementia, or major pulmonary disease, were compared to 545 mid-life controls aged 41–54, who represent a group that is unselected for survival from late-life diseases. Health and lifestyle information, personal and family medical history, and blood samples were collected from all participants. Super-Seniors also underwent four geriatric tests. Results Super-Seniors showed high cognitive (Mini-Mental State Exam mean = 28.3) and functional capacity (Instrumental Activities of Daily Living Scale mean = 21.4), as well as high physical function (Timed Up and Go mean = 12.3 seconds) and low levels of depression (Geriatric Depression Scale mean = 1.5). Super-Seniors were less likely to be current smokers than controls, but the frequency of drinking alcohol was the same in both groups. Super-Seniors were more likely to have 4 or more offspring; controls were more likely to have no children. Female Super-Seniors had a mean age of last fertility 1.9 years older than controls, and were 2.3 times more likely to have had a child at ≥ 40 years. The parents of Super-Seniors had mean ages of deaths of 79.3 years for mothers, and 74.5 years for fathers, each exceeding the life expectancy for their era by a decade. Conclusions Super-Seniors are cognitively and physically high functioning individuals who have evaded major age-related chronic diseases into old age, representing the approximately top 1% for healthspan. The familiality of long lifespan of the parents of Super-Seniors supports the hypothesis that heritable factors contribute to this desirable phenotype.
Collapse
Affiliation(s)
- Julius Halaschek-Wiener
- Canada’s Michael Smith Genome Sciences Centre, British Columbia Cancer Agency (BCCA), Vancouver, British Columbia, Canada
| | - Lauren C. Tindale
- Canada’s Michael Smith Genome Sciences Centre, British Columbia Cancer Agency (BCCA), Vancouver, British Columbia, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Jennifer A. Collins
- Canada’s Michael Smith Genome Sciences Centre, British Columbia Cancer Agency (BCCA), Vancouver, British Columbia, Canada
| | - Stephen Leach
- Canada’s Michael Smith Genome Sciences Centre, British Columbia Cancer Agency (BCCA), Vancouver, British Columbia, Canada
| | - Bruce McManus
- PROOF Centre of Excellence, University of British Columbia, Providence Health Care, Vancouver, British Columbia, Canada
| | - Kenneth Madden
- Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Graydon Meneilly
- Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Nhu D. Le
- Cancer Control Research, BCCA, Vancouver, British Columbia, Canada
| | - Joseph M. Connors
- Centre for Lymphoid Cancer, BCCA, Vancouver, British Columbia, Canada
| | - Angela R. Brooks-Wilson
- Canada’s Michael Smith Genome Sciences Centre, British Columbia Cancer Agency (BCCA), Vancouver, British Columbia, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
- * E-mail:
| |
Collapse
|
73
|
Kulminski AM, Huang J, Loika Y, Arbeev KG, Bagley O, Yashkin A, Duan M, Culminskaya I. Strong impact of natural-selection-free heterogeneity in genetics of age-related phenotypes. Aging (Albany NY) 2018; 10:492-514. [PMID: 29615537 PMCID: PMC5892700 DOI: 10.18632/aging.101407] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 03/24/2018] [Indexed: 11/25/2022]
Abstract
A conceptual difficulty in genetics of age-related phenotypes that make individuals vulnerable to disease in post-reproductive life is genetic heterogeneity attributed to an undefined role of evolution in establishing their molecular mechanisms. Here, we performed univariate and pleiotropic genome-wide meta-analyses of 20 age-related phenotypes leveraging longitudinal information in a sample of 33,431 individuals and dealing with the natural-selection-free genetic heterogeneity. We identified 142 non-proxy single nucleotide polymorphisms (SNPs) with phenotype-specific (18 SNPs) and pleiotropic (124 SNPs) associations at genome-wide level. Univariate meta-analysis identified two novel (11.1%) and replicated 16 SNPs whereas pleiotropic meta-analysis identified 115 novel (92.7%) and nine replicated SNPs. Pleiotropic associations for most novel (93.9%) and all replicated SNPs were strongly impacted by the natural-selection-free genetic heterogeneity in its unconventional form of antagonistic heterogeneity, implying antagonistic directions of genetic effects for directly correlated phenotypes. Our results show that the common genome-wide approach is well adapted to handle homogeneous univariate associations within Mendelian framework whereas most associations with age-related phenotypes are more complex and well beyond that framework. Dissecting the natural-selection-free genetic heterogeneity is critical for gaining insights into genetics of age-related phenotypes and has substantial and unexplored yet potential for improving efficiency of genome-wide analysis.
Collapse
Affiliation(s)
- Alexander M. Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC 27708, USA
| | - Jian Huang
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC 27708, USA
| | - Yury Loika
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC 27708, USA
| | - Konstantin G. Arbeev
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC 27708, USA
| | - Olivia Bagley
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC 27708, USA
| | - Arseniy Yashkin
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC 27708, USA
| | - Matt Duan
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC 27708, USA
| | - Irina Culminskaya
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC 27708, USA
| |
Collapse
|
74
|
Huang H, Zhu Y, Eliot MN, Knopik VS, McGeary JE, Carskadon MA, Hart AC. Combining Human Epigenetics and Sleep Studies in Caenorhabditis elegans: A Cross-Species Approach for Finding Conserved Genes Regulating Sleep. Sleep 2018; 40:3738764. [PMID: 28431118 DOI: 10.1093/sleep/zsx063] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Study Objectives We aimed to test a combined approach to identify conserved genes regulating sleep and to explore the association between DNA methylation and sleep length. Methods We identified candidate genes associated with shorter versus longer sleep duration in college students based on DNA methylation using Illumina Infinium HumanMethylation450 BeadChip arrays. Orthologous genes in Caenorhabditis elegans were identified, and we examined whether their loss of function affected C. elegans sleep. For genes whose perturbation affected C. elegans sleep, we subsequently undertook a small pilot study to re-examine DNA methylation in an independent set of human participants with shorter versus longer sleep durations. Results Eighty-seven out of 485,577 CpG sites had significant differential methylation in young adults with shorter versus longer sleep duration, corresponding to 52 candidate genes. We identified 34 C. elegans orthologs, including NPY/flp-18 and flp-21, which are known to affect sleep. Loss of five additional genes alters developmentally timed C. elegans sleep (B4GALT6/bre-4, DOCK180/ced-5, GNB2L1/rack-1, PTPRN2/ida-1, ZFYVE28/lst-2). For one of these genes, ZFYVE28 (also known as hLst2), the pilot replication study again found decreased DNA methylation associated with shorter sleep duration at the same two CpG sites in the first intron of ZFYVE28. Conclusions Using an approach that combines human epigenetics and C. elegans sleep studies, we identified five genes that play previously unidentified roles in C. elegans sleep. We suggest sleep duration in humans may be associated with differential DNA methylation at specific sites and that the conserved genes identified here likely play roles in C. elegans sleep and in other species.
Collapse
Affiliation(s)
- Huiyan Huang
- Department of Neuroscience, Brown University, Providence, RI
| | - Yong Zhu
- Department of Environmental Health Sciences, Yale University School of Public Health, New Haven, CT
| | - Melissa N Eliot
- Department of Epidemiology, Brown University, Providence, RI
| | - Valerie S Knopik
- Division of Behavioral Genetics, Department of Psychiatry, Rhode Island Hospital, Providence, RI.,Department of Psychiatry and Human Behavior, Brown University, Providence, RI
| | - John E McGeary
- Division of Behavioral Genetics, Department of Psychiatry, Rhode Island Hospital, Providence, RI.,Department of Psychiatry and Human Behavior, Brown University, Providence, RI.,Providence Veterans Affairs Medical Center, Providence, RI
| | - Mary A Carskadon
- Department of Psychiatry and Human Behavior, Brown University, Providence, RI.,E.P. Bradley Hospital Sleep Research Laboratory, Providence, RI.,Center for Sleep Research, University of South Australia, Adelaide, Australia
| | - Anne C Hart
- Department of Neuroscience, Brown University, Providence, RI
| |
Collapse
|
75
|
Ruan L, Zhang X, Li R. Recent insights into the cellular and molecular determinants of aging. J Cell Sci 2018; 131:131/3/jcs210831. [PMID: 29420249 DOI: 10.1242/jcs.210831] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Aging is the gradual decline of physiological functions and organismal fitness, which leads to age-dependent fitness loss, diseases and eventually mortality. Understanding the cause of aging constitutes one of most intriguing areas of research in biology. On both the cellular and molecular levels, it has been hypothesized that there are aging determinants to control the onset and progression of aging, including the loss of beneficial components and accumulation of detrimental factors. This Review highlights the recent advance in identifying various factors that affect the aging process, focusing on how these determinants affect the lifespan and fitness of a cell or organism. With more and more aging determinants revealed, further understanding about their functions and interconnections could enable the development of specific intervention to extend healthy lifespan and reduce the risk of age-related diseases.
Collapse
Affiliation(s)
- Linhao Ruan
- Center for Cell Dynamics, Department of Cell Biology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, USA.,Biochemistry, Cellular and Molecular Biology (BCMB) Graduate Program, Johns Hopkins University School of Medicine, 1830 E. Monument Street, Baltimore, MD 21287, USA
| | - Xi Zhang
- Center for Cell Dynamics, Department of Cell Biology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | - Rong Li
- Center for Cell Dynamics, Department of Cell Biology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, USA .,Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| |
Collapse
|
76
|
Aramillo Irizar P, Schäuble S, Esser D, Groth M, Frahm C, Priebe S, Baumgart M, Hartmann N, Marthandan S, Menzel U, Müller J, Schmidt S, Ast V, Caliebe A, König R, Krawczak M, Ristow M, Schuster S, Cellerino A, Diekmann S, Englert C, Hemmerich P, Sühnel J, Guthke R, Witte OW, Platzer M, Ruppin E, Kaleta C. Transcriptomic alterations during ageing reflect the shift from cancer to degenerative diseases in the elderly. Nat Commun 2018; 9:327. [PMID: 29382830 PMCID: PMC5790807 DOI: 10.1038/s41467-017-02395-2] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 11/27/2017] [Indexed: 02/07/2023] Open
Abstract
Disease epidemiology during ageing shows a transition from cancer to degenerative chronic disorders as dominant contributors to mortality in the old. Nevertheless, it has remained unclear to what extent molecular signatures of ageing reflect this phenomenon. Here we report on the identification of a conserved transcriptomic signature of ageing based on gene expression data from four vertebrate species across four tissues. We find that ageing-associated transcriptomic changes follow trajectories similar to the transcriptional alterations observed in degenerative ageing diseases but are in opposite direction to the transcriptomic alterations observed in cancer. We confirm the existence of a similar antagonism on the genomic level, where a majority of shared risk alleles which increase the risk of cancer decrease the risk of chronic degenerative disorders and vice versa. These results reveal a fundamental trade-off between cancer and degenerative ageing diseases that sheds light on the pronounced shift in their epidemiology during ageing. Ageing is associated with a pronounced shift in mortality from cancer to degenerative diseases. Here, the authors show that in concordance with this shift, conserved transcriptional alterations during ageing across four vertebrates align with degenerative diseases but are opposite to those in cancer.
Collapse
Affiliation(s)
- Peer Aramillo Irizar
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Christian-Albrechts-University Kiel, D-24105, Kiel, Germany
| | - Sascha Schäuble
- Jena University Language and Information Engineering Lab, Friedrich-Schiller-University Jena, D-07743, Jena, Germany.,GerontoSys JenAge Consortium, D-07745, Jena, Germany
| | - Daniela Esser
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Christian-Albrechts-University Kiel, D-24105, Kiel, Germany
| | - Marco Groth
- GerontoSys JenAge Consortium, D-07745, Jena, Germany.,Genome Analysis Lab, Leibniz Institute on Aging-Fritz-Lipmann-Institute, D-07745, Jena, Germany
| | - Christiane Frahm
- GerontoSys JenAge Consortium, D-07745, Jena, Germany.,Hans Berger Department of Neurology, Jena University Hospital, D-07747, Jena, Germany
| | - Steffen Priebe
- GerontoSys JenAge Consortium, D-07745, Jena, Germany.,Systems Biology and Bioinformatics Group, Leibniz Institute for Natural Product Research and Infection Biology-Hans-Knöll-Institute, D-07745, Jena, Germany
| | - Mario Baumgart
- GerontoSys JenAge Consortium, D-07745, Jena, Germany.,Biology of Ageing Lab, Leibniz Institute on Aging-Fritz-Lipmann-Institute, D-07745, Jena, Germany
| | - Nils Hartmann
- GerontoSys JenAge Consortium, D-07745, Jena, Germany.,Molecular Genetics Lab, Leibniz Institute on Aging-Fritz-Lipmann-Institute, D-07745, Jena, Germany
| | - Shiva Marthandan
- GerontoSys JenAge Consortium, D-07745, Jena, Germany.,Imageing Facility, Leibniz Institute on Aging-Fritz-Lipmann-Institute, D-07745, Jena, Germany
| | - Uwe Menzel
- GerontoSys JenAge Consortium, D-07745, Jena, Germany.,Systems Biology and Bioinformatics Group, Leibniz Institute for Natural Product Research and Infection Biology-Hans-Knöll-Institute, D-07745, Jena, Germany
| | - Jule Müller
- Hans Berger Department of Neurology, Jena University Hospital, D-07747, Jena, Germany
| | - Silvio Schmidt
- GerontoSys JenAge Consortium, D-07745, Jena, Germany.,Hans Berger Department of Neurology, Jena University Hospital, D-07747, Jena, Germany
| | - Volker Ast
- Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital, D-07747, Jena, Germany.,Network Modeling, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute, D-07745, Jena, Germany
| | - Amke Caliebe
- Institute for Medical Informatics and Statistics, Christian-Albrechts-University Kiel, D-24105, Kiel, Germany
| | - Rainer König
- Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena University Hospital, D-07747, Jena, Germany.,Network Modeling, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute, D-07745, Jena, Germany
| | - Michael Krawczak
- Institute for Medical Informatics and Statistics, Christian-Albrechts-University Kiel, D-24105, Kiel, Germany
| | - Michael Ristow
- GerontoSys JenAge Consortium, D-07745, Jena, Germany.,Energy Metabolism Laboratory, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach/Zürich, CH-8603, Switzerland
| | - Stefan Schuster
- GerontoSys JenAge Consortium, D-07745, Jena, Germany.,Department of Bioinformatics, Friedrich-Schiller-University Jena, D-07743, Jena, Germany
| | - Alessandro Cellerino
- GerontoSys JenAge Consortium, D-07745, Jena, Germany.,Biology of Ageing Lab, Leibniz Institute on Aging-Fritz-Lipmann-Institute, D-07745, Jena, Germany.,Laboratory of Neurobiology, Scuola Normale Superiore, University of Pisa, I-56100, Pisa, Italy
| | - Stephan Diekmann
- GerontoSys JenAge Consortium, D-07745, Jena, Germany.,Molecular Biology Lab, Leibniz Institute on Aging-Fritz-Lipmann-Institute, D-07745, Jena, Germany
| | - Christoph Englert
- GerontoSys JenAge Consortium, D-07745, Jena, Germany.,Molecular Genetics Lab, Leibniz Institute on Aging-Fritz-Lipmann-Institute, D-07745, Jena, Germany.,Faculty of Biology and Pharmacy, Friedrich-Schiller-University Jena, D-07743, Jena, Germany
| | - Peter Hemmerich
- GerontoSys JenAge Consortium, D-07745, Jena, Germany.,Imageing Facility, Leibniz Institute on Aging-Fritz-Lipmann-Institute, D-07745, Jena, Germany
| | - Jürgen Sühnel
- GerontoSys JenAge Consortium, D-07745, Jena, Germany.,Biocomputing Lab, Leibniz Institute on Aging-Fritz-Lipmann-Institute, D-07745, Jena, Germany
| | - Reinhard Guthke
- GerontoSys JenAge Consortium, D-07745, Jena, Germany.,Systems Biology and Bioinformatics Group, Leibniz Institute for Natural Product Research and Infection Biology-Hans-Knöll-Institute, D-07745, Jena, Germany
| | - Otto W Witte
- GerontoSys JenAge Consortium, D-07745, Jena, Germany.,Hans Berger Department of Neurology, Jena University Hospital, D-07747, Jena, Germany
| | - Matthias Platzer
- GerontoSys JenAge Consortium, D-07745, Jena, Germany.,Genome Analysis Lab, Leibniz Institute on Aging-Fritz-Lipmann-Institute, D-07745, Jena, Germany
| | - Eytan Ruppin
- Department of Computer Science and Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD, 20742, USA
| | - Christoph Kaleta
- Research Group Medical Systems Biology, Institute of Experimental Medicine, Christian-Albrechts-University Kiel, D-24105, Kiel, Germany. .,GerontoSys JenAge Consortium, D-07745, Jena, Germany.
| |
Collapse
|
77
|
Latendresse SJ, Musci R, Maher BS. Critical Issues in the Inclusion of Genetic and Epigenetic Information in Prevention and Intervention Trials. PREVENTION SCIENCE : THE OFFICIAL JOURNAL OF THE SOCIETY FOR PREVENTION RESEARCH 2018; 19:58-67. [PMID: 28409280 PMCID: PMC5640466 DOI: 10.1007/s11121-017-0785-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Human genetic research in the past decade has generated a wealth of data from the genome-wide association scan era, much of which is catalogued and freely available. These data will typically test the relationship between a single nucleotide variant or polymorphism (SNP) and some outcome, disease, or trait. Ongoing investigations will yield a similar wealth of data regarding epigenetic phenomena. These data will typically test the relationship between DNA methylation at a single genomic location/region and some outcome. Most of these findings will be the result of cross-sectional investigations typically using ascertained cases and controls. Consequently, most methodological consideration focuses on methods appropriate for simple case-control comparisons. It is expected that a growing number of investigators with longitudinal experimental prevention or intervention cohorts will also measure genetic and epigenetic indicators as part of their investigations, harvesting the wealth of information generated by the genome-wide association study (GWAS) era to allow for targeted hypothesis testing in the next generation of prevention and intervention trials. Herein, we discuss appropriate quality control and statistical modelling of genetic, polygenic, and epigenetic measures in longitudinal models. We specifically discuss quality control, population stratification, genotype imputation, pathway approaches, and proper modelling of an interaction between a specific genetic variant and an environment variable (GxE interaction).
Collapse
Affiliation(s)
- Shawn J Latendresse
- Department of Psychology and Neuroscience, Baylor University, One Bear Place #97334, Waco, TX, 76798, USA.
| | - Rashelle Musci
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, 624 N. Broadway Ave, Baltimore, MD, 21205, USA
| | - Brion S Maher
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, 624 N. Broadway Ave, Baltimore, MD, 21205, USA.
| |
Collapse
|
78
|
Yin Y, Wang Z. ApoE and Neurodegenerative Diseases in Aging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1086:77-92. [PMID: 30232753 DOI: 10.1007/978-981-13-1117-8_5] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Age and apolipoprotein E (ApoE) are the mightiest risk factors for dementia and cardiovascular diseases, but the underlying mechanisms remain unclear. In human, ApoE has three isoforms, ApoE2, ApoE3, and ApoE4, which are expressed by the polymorphic alleles: ɛ2, ɛ3, and ɛ4. Among the three polymorphic alleles, apoE ε4 is the most risk gene. ApoE is the main ligand for the low-density lipoprotein (LDL) receptor and the LDL receptor-related protein (LRP), functioning as the component of plasma lipoproteins in the transportation of lipids. Physiologically, ApoE is a multifunctional protein with central roles in lipid metabolism; it transports lipids, including cholesterol, through the cerebrospinal fluid (CSF) and plasma. ApoE expression regulation and apoE gene polymorphism have an important connection with neurological or neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), ischemic stroke, and other diseases.
Collapse
Affiliation(s)
- Yuemiao Yin
- MOE Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Zhao Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
79
|
Abstract
Cancer is largely an aging disease. Accelerated biological aging may be the strongest predictor of cancer and other chronic disease risks. In the absence of reliable and quantifiable biomarkers of aging to date, it has long been observed that tumorigenesis shares distinct epigenetic alterations with the aging process. Recently, epigenetic age estimates have been developed based on the availability of genome-wide DNA methylation profiles, by applying in the prediction formula the methylation level at a subset of highly predictive methylation sites, called epigenetic clock. These DNA methylation age estimates have produced remarkably strong correlations with chronological age, with a small deviation and high reproducibility across different age groups and study populations. Moreover, an increasing number of epidemiologic studies have demonstrated an independent association of DNA methylation age or the extent of acceleration with mortality and various aging-related conditions, even after accounting for differences in chronological age and other risk factors. Although epigenetic profiles are known to be tissue-specific, both target tissue- and multiple tissue-derived estimates appear to perform well to capture what is thought to be the cumulative epigenetic drift that represents a multifactorial degenerative process across tissues and organisms. Further refinement of the epigenetic age estimates is anticipated over time to accommodate a better technological coverage of the methylome and a better understanding of the biology underlying predictive regions. Epidemiologic principles will remain critical for the evaluation of research findings involving, for example, different study populations, design, follow-up time, and quality of covariate data. Overall, the epigenetic age estimates are an exciting development with useful implications for biomedical research of healthy aging and disease prevention and control.
Collapse
|
80
|
Yanai H, Fraifeld VE. The role of cellular senescence in aging through the prism of Koch-like criteria. Ageing Res Rev 2018; 41:18-33. [PMID: 29106993 DOI: 10.1016/j.arr.2017.10.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 09/11/2017] [Accepted: 10/23/2017] [Indexed: 12/13/2022]
Abstract
Since Hayflick's discovery of cellular senescence (CS), a great volume of knowledge in the field has been accumulated and intensively discussed. Here, we attempted to organize the evidence "for" and "against" the hypothesized causal role of CS in aging. For that purpose, we utilized robust Koch-like logical criteria, based on the assumption that some quantitative relationships between the accumulation of senescent cells and aging rate should exist. If so, it could be expected that (i) the "CS load" would be greater in the premature aging phenotype and lesser in longevity phenotype; (ii) CS would promote age-related diseases, and (iii) the interventions that modulate the levels of senescent cells should also modulate health/lifespan. The analysis shows that CS can be considered a causal factor of aging and an important player in various age-related diseases, though its contribution may greatly vary across species. While the relative impact of senescent cells to aging could overall be rather limited and their elimination is hardly expected to be the "fountain of youth", the potential benefits of the senolytic strategy seems a promising option in combating age-related diseases and extending healthspan.
Collapse
|
81
|
Santoro A, Ostan R, Candela M, Biagi E, Brigidi P, Capri M, Franceschi C. Gut microbiota changes in the extreme decades of human life: a focus on centenarians. Cell Mol Life Sci 2018; 75:129-148. [PMID: 29032502 PMCID: PMC5752746 DOI: 10.1007/s00018-017-2674-y] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 09/29/2017] [Indexed: 12/20/2022]
Abstract
The gut microbiota (GM) is a complex, evolutionarily molded ecological system, which contributes to a variety of physiological functions. The GM is highly dynamic, being sensitive to environmental stimuli, and its composition changes over the host's entire lifespan. However, the basic question of how much these changes may be ascribed to variables such as population, diet, genetics and gender, and/or to the aging process per se is still largely unanswered. We argue that comparison among studies on centenarians-the best model of healthy aging and longevity-recruited from different geographical areas/populations (different genetics and dietary habits) can help to disentangle the contribution of aging and non-aging-related variables to GM remodeling with age. The current review focuses on the role of population, gender and host genetics as possible drivers of GM modification along the human aging process. The feedback impact of age-associated GM variation on the GM-brain axis and GM metabolomics is also discussed. We likewise address the role of GM in neurodegenerative diseases such as Parkinson's and Alzheimer's, and its possible therapeutic use, taking advantage of the fact that centenarians are characterized by an extreme (healthy) phenotype versus patients suffering from age-related pathologies. Finally, it is argued that longitudinal studies combining metagenomics sequencing and in-depth phylogenetic analysis with a comprehensive phenotypic characterization of centenarians and patients using up-to-date omics (metabolomics, transcriptomics and meta-transcriptomics) are urgently needed.
Collapse
Affiliation(s)
- Aurelia Santoro
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum-University of Bologna, Via San Giacomo 12, 40126, Bologna, Italy.
- Interdepartmental Centre "L. Galvani" (CIG) Alma Mater Studiorum-University of Bologna, Via San Giacomo 12, 40126, Bologna, Italy.
| | - Rita Ostan
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum-University of Bologna, Via San Giacomo 12, 40126, Bologna, Italy
- Interdepartmental Centre "L. Galvani" (CIG) Alma Mater Studiorum-University of Bologna, Via San Giacomo 12, 40126, Bologna, Italy
| | - Marco Candela
- Department of Pharmacy and Biotechnology (FABIT), Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy
| | - Elena Biagi
- Department of Pharmacy and Biotechnology (FABIT), Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy
| | - Patrizia Brigidi
- Department of Pharmacy and Biotechnology (FABIT), Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy
| | - Miriam Capri
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum-University of Bologna, Via San Giacomo 12, 40126, Bologna, Italy
- Interdepartmental Centre "L. Galvani" (CIG) Alma Mater Studiorum-University of Bologna, Via San Giacomo 12, 40126, Bologna, Italy
| | - Claudio Franceschi
- Institute of Neurological Sciences (IRCCS), Via Altura 3, 40139, Bologna, Italy
| |
Collapse
|
82
|
Pilling LC, Kuo CL, Sicinski K, Tamosauskaite J, Kuchel GA, Harries LW, Herd P, Wallace R, Ferrucci L, Melzer D. Human longevity: 25 genetic loci associated in 389,166 UK biobank participants. Aging (Albany NY) 2017; 9:2504-2520. [PMID: 29227965 PMCID: PMC5764389 DOI: 10.18632/aging.101334] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 11/26/2017] [Indexed: 12/22/2022]
Abstract
We undertook a genome-wide association study (GWAS) of parental longevity in European descent UK Biobank participants. For combined mothers' and fathers' attained age, 10 loci were associated (p<5*10-8), including 8 previously identified for traits including survival, Alzheimer's and cardiovascular disease. Of these, 4 were also associated with longest 10% survival (mothers age ≥90 years, fathers ≥87 years), with 2 additional associations including MC2R intronic variants (coding for the adrenocorticotropic hormone receptor). Mother's age at death was associated with 3 additional loci (2 linked to autoimmune conditions), and 8 for fathers only. An attained age genetic risk score associated with parental survival in the US Health and Retirement Study and the Wisconsin Longitudinal Study and with having a centenarian parent (n=1,181) in UK Biobank. The results suggest that human longevity is highly polygenic with prominent roles for loci likely involved in cellular senescence and inflammation, plus lipid metabolism and cardiovascular conditions. There may also be gender specific routes to longevity.
Collapse
Affiliation(s)
- Luke C. Pilling
- Epidemiology and Public Health Group, University of Exeter Medical School, RILD Level 3, Royal Devon & Exeter Hospital, Exeter, EX2 5DW, UK
| | - Chia-Ling Kuo
- Department of Community Medicine and Health Care, Connecticut Institute for Clinical and Translational Science, Institute for Systems Genomics, University of Connecticut Health Center, CT 06269 USA
| | - Kamil Sicinski
- Center for Demography of Health and Aging, University of Wisconsin, Madison, WI 53706, USA
| | - Jone Tamosauskaite
- Epidemiology and Public Health Group, University of Exeter Medical School, RILD Level 3, Royal Devon & Exeter Hospital, Exeter, EX2 5DW, UK
| | - George A. Kuchel
- UConn Center on Aging, University of Connecticut, Farmington, CT 06030, USA
| | - Lorna W. Harries
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, RILD Level 3, Royal Devon & Exeter Hospital, Exeter, UK
| | - Pamela Herd
- La Follette School of Public Affairs and the Department of Sociology, University of Wisconsin, Madison, WI 53706, USA
| | - Robert Wallace
- College of Public Health, University of Iowa, Iowa City, IA 52242, USA
| | | | - David Melzer
- Epidemiology and Public Health Group, University of Exeter Medical School, RILD Level 3, Royal Devon & Exeter Hospital, Exeter, EX2 5DW, UK
- UConn Center on Aging, University of Connecticut, Farmington, CT 06030, USA
| |
Collapse
|
83
|
Pan C, McInnes G, Deflaux N, Snyder M, Bingham J, Datta S, Tsao PS. Cloud-based interactive analytics for terabytes of genomic variants data. Bioinformatics 2017; 33:3709-3715. [PMID: 28961771 PMCID: PMC5860318 DOI: 10.1093/bioinformatics/btx468] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 06/30/2017] [Accepted: 07/25/2017] [Indexed: 12/30/2022] Open
Abstract
MOTIVATION Large scale genomic sequencing is now widely used to decipher questions in diverse realms such as biological function, human diseases, evolution, ecosystems, and agriculture. With the quantity and diversity these data harbor, a robust and scalable data handling and analysis solution is desired. RESULTS We present interactive analytics using a cloud-based columnar database built on Dremel to perform information compression, comprehensive quality controls, and biological information retrieval in large volumes of genomic data. We demonstrate such Big Data computing paradigms can provide orders of magnitude faster turnaround for common genomic analyses, transforming long-running batch jobs submitted via a Linux shell into questions that can be asked from a web browser in seconds. Using this method, we assessed a study population of 475 deeply sequenced human genomes for genomic call rate, genotype and allele frequency distribution, variant density across the genome, and pharmacogenomic information. AVAILABILITY AND IMPLEMENTATION Our analysis framework is implemented in Google Cloud Platform and BigQuery. Codes are available at https://github.com/StanfordBioinformatics/mvp_aaa_codelabs. CONTACT cuiping@stanford.edu or ptsao@stanford.edu. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Cuiping Pan
- VA Palo Alto Health Care System, Palo Alto Epidemiology Research and Information Center for Genomics, CA, USA
- Department of Genetics, Stanford University, CA, USA
| | - Gregory McInnes
- VA Palo Alto Health Care System, Palo Alto Epidemiology Research and Information Center for Genomics, CA, USA
- Stanford Center for Genomics and Personalized Medicine, Stanford University, CA, USA
| | - Nicole Deflaux
- Google, Mountain View, CA, USA
- Verily Life Sciences, South San Francisco, CA, USA
| | - Michael Snyder
- Department of Genetics, Stanford University, CA, USA
- Stanford Center for Genomics and Personalized Medicine, Stanford University, CA, USA
| | - Jonathan Bingham
- Google, Mountain View, CA, USA
- Verily Life Sciences, South San Francisco, CA, USA
| | - Somalee Datta
- VA Palo Alto Health Care System, Palo Alto Epidemiology Research and Information Center for Genomics, CA, USA
- Stanford Center for Genomics and Personalized Medicine, Stanford University, CA, USA
| | - Philip S Tsao
- VA Palo Alto Health Care System, Palo Alto Epidemiology Research and Information Center for Genomics, CA, USA
- Division of Cardiovascular Medicine, Stanford University, Stanford, CA, USA
| |
Collapse
|
84
|
Sebastiani P, Gurinovich A, Bae H, Andersen S, Malovini A, Atzmon G, Villa F, Kraja AT, Ben-Avraham D, Barzilai N, Puca A, Perls TT. Four Genome-Wide Association Studies Identify New Extreme Longevity Variants. J Gerontol A Biol Sci Med Sci 2017; 72:1453-1464. [PMID: 28329165 DOI: 10.1093/gerona/glx027] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 02/14/2017] [Indexed: 01/10/2023] Open
Abstract
The search for the genetic determinants of extreme human longevity has been challenged by the phenotype's rarity and its nonspecific definition by investigators. To address these issues, we established a consortium of four studies of extreme longevity that contributed 2,070 individuals who survived to the oldest one percentile of survival for the 1900 U.S. birth year cohort. We conducted various analyses to discover longevity-associated variants (LAV) and characterized those LAVs that differentiate survival to extreme age at death (eSAVs) from those LAVs that become more frequent in centenarians because of mortality selection (eg, survival to younger years). The analyses identified new rare variants in chromosomes 4 and 7 associated with extreme survival and with reduced risk for cardiovascular disease and Alzheimer's disease. The results confirm the importance of studying truly rare survival to discover those combinations of common and rare variants associated with extreme longevity and longer health span.
Collapse
Affiliation(s)
- Paola Sebastiani
- Department of Biostatistics, Boston University School of Public Health, Massachusetts
| | | | - Harold Bae
- College of Public Health and Human Sciences, Oregon State University, Corvallis
| | - Stacy Andersen
- Geriatrics Section, Department of Medicine, Boston University School of Medicine & Boston Medical Center, Massachusetts
| | - Alberto Malovini
- Laboratory of Informatics and Systems Engineering for Clinical Research, IRCCS Fondazione Salvatore Maugeri, Pavia, Italy
| | - Gil Atzmon
- Department of Natural Science, University of Haifa, Israel.,Department of Medicine.,Department of Genetics, Albert Einstein College of Medicine, Bronx, New York
| | - Francesco Villa
- IRCCS MultiMedica, Milan, Italy.,Department of Medicine and Surgery, University of Salerno, Baronissi, Italy
| | - Aldi T Kraja
- Division of Statistical Genomics, Washington University School of Medicine, Saint Louis, Missouri
| | - Danny Ben-Avraham
- Department of Medicine.,Department of Genetics, Albert Einstein College of Medicine, Bronx, New York
| | - Nir Barzilai
- Department of Medicine.,Department of Genetics, Albert Einstein College of Medicine, Bronx, New York
| | - Annibale Puca
- IRCCS MultiMedica, Milan, Italy.,Department of Medicine and Surgery, University of Salerno, Baronissi, Italy
| | - Thomas T Perls
- Geriatrics Section, Department of Medicine, Boston University School of Medicine & Boston Medical Center, Massachusetts
| |
Collapse
|
85
|
Joshi PK, Pirastu N, Kentistou KA, Fischer K, Hofer E, Schraut KE, Clark DW, Nutile T, Barnes CLK, Timmers PRHJ, Shen X, Gandin I, McDaid AF, Hansen TF, Gordon SD, Giulianini F, Boutin TS, Abdellaoui A, Zhao W, Medina-Gomez C, Bartz TM, Trompet S, Lange LA, Raffield L, van der Spek A, Galesloot TE, Proitsi P, Yanek LR, Bielak LF, Payton A, Murgia F, Concas MP, Biino G, Tajuddin SM, Seppälä I, Amin N, Boerwinkle E, Børglum AD, Campbell A, Demerath EW, Demuth I, Faul JD, Ford I, Gialluisi A, Gögele M, Graff M, Hingorani A, Hottenga JJ, Hougaard DM, Hurme MA, Ikram MA, Jylhä M, Kuh D, Ligthart L, Lill CM, Lindenberger U, Lumley T, Mägi R, Marques-Vidal P, Medland SE, Milani L, Nagy R, Ollier WER, Peyser PA, Pramstaller PP, Ridker PM, Rivadeneira F, Ruggiero D, Saba Y, Schmidt R, Schmidt H, Slagboom PE, Smith BH, Smith JA, Sotoodehnia N, Steinhagen-Thiessen E, van Rooij FJA, Verbeek AL, Vermeulen SH, Vollenweider P, Wang Y, Werge T, Whitfield JB, Zonderman AB, Lehtimäki T, Evans MK, Pirastu M, Fuchsberger C, Bertram L, Pendleton N, Kardia SLR, Ciullo M, Becker DM, Wong A, Psaty BM, van Duijn CM, Wilson JG, Jukema JW, Kiemeney L, Uitterlinden AG, et alJoshi PK, Pirastu N, Kentistou KA, Fischer K, Hofer E, Schraut KE, Clark DW, Nutile T, Barnes CLK, Timmers PRHJ, Shen X, Gandin I, McDaid AF, Hansen TF, Gordon SD, Giulianini F, Boutin TS, Abdellaoui A, Zhao W, Medina-Gomez C, Bartz TM, Trompet S, Lange LA, Raffield L, van der Spek A, Galesloot TE, Proitsi P, Yanek LR, Bielak LF, Payton A, Murgia F, Concas MP, Biino G, Tajuddin SM, Seppälä I, Amin N, Boerwinkle E, Børglum AD, Campbell A, Demerath EW, Demuth I, Faul JD, Ford I, Gialluisi A, Gögele M, Graff M, Hingorani A, Hottenga JJ, Hougaard DM, Hurme MA, Ikram MA, Jylhä M, Kuh D, Ligthart L, Lill CM, Lindenberger U, Lumley T, Mägi R, Marques-Vidal P, Medland SE, Milani L, Nagy R, Ollier WER, Peyser PA, Pramstaller PP, Ridker PM, Rivadeneira F, Ruggiero D, Saba Y, Schmidt R, Schmidt H, Slagboom PE, Smith BH, Smith JA, Sotoodehnia N, Steinhagen-Thiessen E, van Rooij FJA, Verbeek AL, Vermeulen SH, Vollenweider P, Wang Y, Werge T, Whitfield JB, Zonderman AB, Lehtimäki T, Evans MK, Pirastu M, Fuchsberger C, Bertram L, Pendleton N, Kardia SLR, Ciullo M, Becker DM, Wong A, Psaty BM, van Duijn CM, Wilson JG, Jukema JW, Kiemeney L, Uitterlinden AG, Franceschini N, North KE, Weir DR, Metspalu A, Boomsma DI, Hayward C, Chasman D, Martin NG, Sattar N, Campbell H, Esko T, Kutalik Z, Wilson JF. Genome-wide meta-analysis associates HLA-DQA1/DRB1 and LPA and lifestyle factors with human longevity. Nat Commun 2017; 8:910. [PMID: 29030599 PMCID: PMC5715013 DOI: 10.1038/s41467-017-00934-5] [Show More Authors] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 08/08/2017] [Indexed: 01/03/2023] Open
Abstract
Genomic analysis of longevity offers the potential to illuminate the biology of human aging. Here, using genome-wide association meta-analysis of 606,059 parents’ survival, we discover two regions associated with longevity (HLA-DQA1/DRB1 and LPA). We also validate previous suggestions that APOE, CHRNA3/5, CDKN2A/B, SH2B3 and FOXO3A influence longevity. Next we show that giving up smoking, educational attainment, openness to new experience and high-density lipoprotein (HDL) cholesterol levels are most positively genetically correlated with lifespan while susceptibility to coronary artery disease (CAD), cigarettes smoked per day, lung cancer, insulin resistance and body fat are most negatively correlated. We suggest that the effect of education on lifespan is principally mediated through smoking while the effect of obesity appears to act via CAD. Using instrumental variables, we suggest that an increase of one body mass index unit reduces lifespan by 7 months while 1 year of education adds 11 months to expected lifespan. Variability in human longevity is genetically influenced. Using genetic data of parental lifespan, the authors identify associations at HLA-DQA/DRB1 and LPA and find that genetic variants that increase educational attainment have a positive effect on lifespan whereas increasing BMI negatively affects lifespan.
Collapse
Affiliation(s)
- Peter K Joshi
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH8 9AG, UK.
| | - Nicola Pirastu
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH8 9AG, UK
| | - Katherine A Kentistou
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH8 9AG, UK.,Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, Scotland
| | - Krista Fischer
- Estonian Genome Center, University of Tartu, University of Tartu, Tartu, 51010, Estonia
| | - Edith Hofer
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Graz, 8036, Austria.,Institute of Medical Informatics, Statistics and Documentation, Medical University of Graz, Graz, 8036, Austria
| | - Katharina E Schraut
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH8 9AG, UK.,Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, Scotland
| | - David W Clark
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH8 9AG, UK
| | - Teresa Nutile
- Institute of Genetics and Biophysics "A. Buzzati-Traverso" - CNR, Naples, 80131, Italy
| | - Catriona L K Barnes
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH8 9AG, UK
| | - Paul R H J Timmers
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH8 9AG, UK
| | - Xia Shen
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH8 9AG, UK.,Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | - Ilaria Gandin
- Department of Medical Sciences, University of Trieste, Trieste, 34100, Italy.,Institute for Maternal and Child Health, IRCCS "Burlo Garofolo", Trieste, 34137, Italy
| | - Aaron F McDaid
- Institute of Social and Preventive Medicine, Lausanne University Hospital, Lausanne, 1010, Switzerland.,Swiss Institute of Bioinformatics, Lausanne, 1015, Switzerland
| | - Thomas Folkmann Hansen
- Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, Mental Health Services Copenhagen, Roskilde, DK-4000, Denmark.,iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, DK-8000, Denmark
| | - Scott D Gordon
- QIMR Berghofer Institute of Medical Research, Brisbane, QLD, 4006, Australia
| | - Franco Giulianini
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, 02215, USA
| | - Thibaud S Boutin
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Abdel Abdellaoui
- Netherlands Twin Register, Department of Biological Psychology, Vrije Universiteit, Amsterdam, Amsterdam Public Health Institute (APH), Amsterdam, 1081BT, Netherlands
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Carolina Medina-Gomez
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, 3015 CN, Netherlands.,Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015 CN, Netherlands
| | - Traci M Bartz
- Cardiovascular Health Research Unit, Departments of Biostatistics and Medicine, University of Washington, Seattle, WA, 98101, USA
| | - Stella Trompet
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, 2300RC, The Netherlands.,Department of Cardiology, Leiden University Medical Center, Leiden, 2300RC, The Netherlands
| | - Leslie A Lange
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Laura Raffield
- Department of Genetics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Ashley van der Spek
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015 CN, Netherlands
| | - Tessel E Galesloot
- Department for Health Evidence, Radboud Institute for Health Sciences, Radboud university medical center, Nijmegen, 6500 HB, The Netherlands
| | - Petroula Proitsi
- MRC Unit for Lifelong Health & Ageing at UCL, University College London, London, WC1B 5JU, UK
| | - Lisa R Yanek
- Department of Medicine, GeneSTAR Research Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Lawrence F Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Antony Payton
- Centre for Epidemiology, Division of Population Health, Health Services Research & Primary Care, The University of Manchester, Manchester, Greater, Manchester, M13 9PL, UK
| | - Federico Murgia
- Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC), (Affiliated Institute of the University of Lübeck, Lübeck, Germany), Bolzano, 39100, Italy
| | - Maria Pina Concas
- Institute of Genetic and Biomedical Research - Support Unity, National Research Council of Italy, Sassari, 07100, Italy
| | - Ginevra Biino
- Institute of Molecular Genetics, National Research Council of Italy, Pavia, 27100, Italy
| | - Salman M Tajuddin
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore City, MD, 21224, USA
| | - Ilkka Seppälä
- Department of Clinical Chemistry, Fimlab Laboratories and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, 33014, Finland
| | - Najaf Amin
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015 CN, Netherlands
| | - Eric Boerwinkle
- Health Science Center at Houston, UTHealth School of Public Health, University of Texas, Houston, TX, 77030, USA
| | - Anders D Børglum
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, DK-8000, Denmark.,Department of Biomedicine-Human Genetics, Aarhus University, DK-8000, Aarhus C, Denmark.,Centre for Integrative Sequencing, iSEQ, Aarhus University, DK-8000, Aarhus C, Denmark
| | - Archie Campbell
- Centre for Genomic & Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Ellen W Demerath
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, 55454, USA
| | - Ilja Demuth
- Charité Research Group on Geriatrics, Charité, Universitätsmedizin Berlin, Berlin, 13347, Germany.,Lipid Clinic at the Interdisciplinary Metabolism Center, Charité, Universitätsmedizin Berlin, Berlin, 13353, Germany.,Institute for Medical and Human Genetics, Charité, Universitätsmedizin Berlin, Berlin, 13353, Germany
| | - Jessica D Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, 48014, USA
| | - Ian Ford
- Robertson Center for biostatistics, University of Glasgow, Glasgow, G12 8QQ, UK
| | | | - Martin Gögele
- Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC), (Affiliated Institute of the University of Lübeck, Lübeck, Germany), Bolzano, 39100, Italy
| | - MariaElisa Graff
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, 27514, USA
| | - Aroon Hingorani
- Institute of Cardiovascular Science, University College London, London, WC1E 6BT, UK
| | - Jouke-Jan Hottenga
- Netherlands Twin Register, Department of Biological Psychology, Vrije Universiteit, Amsterdam, Amsterdam Public Health Institute (APH), Amsterdam, 1081BT, Netherlands
| | - David M Hougaard
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, DK-8000, Denmark.,Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, 2300, Denmark
| | - Mikko A Hurme
- Department of Microbiology and Immunology, Fimlab Laboratories and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, 33014, Finland
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015 CN, Netherlands
| | - Marja Jylhä
- Gerontology Research Center, Tampere, Finland, Faculty of Social Sciences, University of Tampere, Tampere, 33104, Finland
| | - Diana Kuh
- MRC Unit for Lifelong Health & Ageing at UCL, University College London, London, WC1B 5JU, UK
| | - Lannie Ligthart
- Netherlands Twin Register, Department of Biological Psychology, Vrije Universiteit, Amsterdam, Amsterdam Public Health Institute (APH), Amsterdam, 1081BT, Netherlands
| | - Christina M Lill
- Genetic and Molecular Epidemiology Group, Institute of Neurogenetics, University of Lübeck, 23562, Lübeck, Germany
| | - Ulman Lindenberger
- Center for Lifespan Psychology, Max Planck Institute for Human Development, Berlin, 14195, Germany.,Max Planck UCL Centre for Computational Psychiatry and Ageing Research, Berlin, 14195, Germany
| | - Thomas Lumley
- Department of Statistics, University of Auckland, Auckland, 1010, New Zealand
| | - Reedik Mägi
- Estonian Genome Center, University of Tartu, University of Tartu, Tartu, 51010, Estonia
| | - Pedro Marques-Vidal
- Department of Medicine, Internal Medicine, Lausanne University Hospital, Lausanne, 1011, Switzerland
| | - Sarah E Medland
- QIMR Berghofer Institute of Medical Research, Brisbane, QLD, 4006, Australia
| | - Lili Milani
- Estonian Genome Center, University of Tartu, University of Tartu, Tartu, 51010, Estonia
| | - Reka Nagy
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
| | - William E R Ollier
- Division of Population Health, Health Services Research & Primary Care, School of Health Sciences, Manchester Academic Health Science Centre, University of Manchester, Manchester, Greater Manchester, M13 9PL, UK
| | - Patricia A Peyser
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Peter P Pramstaller
- Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC), (Affiliated Institute of the University of Lübeck, Lübeck, Germany), Bolzano, 39100, Italy
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, 02215, USA.,TH Chan School of Public Health, Harvard Medical School, Boston, MA, 02115, USA
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, 3015 CN, Netherlands.,Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015 CN, Netherlands
| | - Daniela Ruggiero
- Institute of Genetics and Biophysics "A. Buzzati-Traverso" - CNR, Naples, 80131, Italy
| | - Yasaman Saba
- Austrian Stroke Prevention Study, Institute of Molecular Biology and Biochemistry, Centre for Molecular Medicine, Medical University of Graz, Graz, 8010, Austria
| | - Reinhold Schmidt
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Graz, 8036, Austria
| | - Helena Schmidt
- Austrian Stroke Prevention Study, Institute of Molecular Biology and Biochemistry, Centre for Molecular Medicine, Medical University of Graz, Graz, 8010, Austria
| | - P Eline Slagboom
- Section of Molecular Epidemiology, Department of medical statistics, Leiden University Medical Center, Leiden, 2300RC, The Netherlands
| | - Blair H Smith
- Division of Population Health Sciences, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109, USA.,Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, 48014, USA
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Division of Cardiology, University of Washington, Seattle, WA, 98101, USA
| | | | - Frank J A van Rooij
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015 CN, Netherlands
| | - André L Verbeek
- Department for Health Evidence, Radboud Institute for Health Sciences, Radboud university medical center, Nijmegen, 6500 HB, The Netherlands
| | - Sita H Vermeulen
- Department for Health Evidence, Radboud Institute for Health Sciences, Radboud university medical center, Nijmegen, 6500 HB, The Netherlands
| | - Peter Vollenweider
- Department of Medicine, Internal Medicine, Lausanne University Hospital, Lausanne, 1011, Switzerland
| | - Yunpeng Wang
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, DK-8000, Denmark.,NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, 0450, Norway
| | - Thomas Werge
- Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, Mental Health Services Copenhagen, Roskilde, DK-4000, Denmark.,iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, DK-8000, Denmark
| | - John B Whitfield
- QIMR Berghofer Institute of Medical Research, Brisbane, QLD, 4006, Australia
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore City, MD, 21224, USA
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, 33014, Finland
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore City, MD, 21224, USA
| | - Mario Pirastu
- Institute of Genetic and Biomedical Research - Support Unity, National Research Council of Italy, Sassari, 07100, Italy
| | - Christian Fuchsberger
- Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC), (Affiliated Institute of the University of Lübeck, Lübeck, Germany), Bolzano, 39100, Italy
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics, Institutes of Neurogenetics & Cardiogenetics, University of Lübeck, Lübeck, 23562, Germany.,Neuroepidemiology and Ageing Research Group, School of Public Health, Imperial College, London, W6 8RP, UK
| | - Neil Pendleton
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Manchester Academic Health Science Centre, University of Manchester, Manchester, Greater Manchester, M13 9PL, UK
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Marina Ciullo
- Institute of Genetics and Biophysics "A. Buzzati-Traverso" - CNR, Naples, 80131, Italy.,IRCCS Neuromed, Pozzilli (IS), 86077, Italy
| | - Diane M Becker
- Department of Medicine, GeneSTAR Research Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Andrew Wong
- MRC Unit for Lifelong Health & Ageing at UCL, University College London, London, WC1B 5JU, UK
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Departments of Epidemiology, Medicine and Health Services, University of Washington, Seattle, WA, 98101, USA.,Kaiser Permanente Washington Health Research Institute, Seattle, WA, 98101, USA
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015 CN, Netherlands
| | - James G Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, 2300RC, The Netherlands
| | - Lambertus Kiemeney
- Department for Health Evidence, Radboud Institute for Health Sciences, Radboud university medical center, Nijmegen, 6500 HB, The Netherlands
| | - André G Uitterlinden
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, 3015 CN, Netherlands.,Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015 CN, Netherlands
| | - Nora Franceschini
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, 27514, USA
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, 27514, USA
| | - David R Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, 48014, USA
| | - Andres Metspalu
- Estonian Genome Center, University of Tartu, University of Tartu, Tartu, 51010, Estonia
| | - Dorret I Boomsma
- Netherlands Twin Register, Department of Biological Psychology, Vrije Universiteit, Amsterdam, Amsterdam Public Health Institute (APH), Amsterdam, 1081BT, Netherlands
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Daniel Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, 02215, USA.,TH Chan School of Public Health, Harvard Medical School, Boston, MA, 02115, USA
| | - Nicholas G Martin
- QIMR Berghofer Institute of Medical Research, Brisbane, QLD, 4006, Australia
| | - Naveed Sattar
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, G12 8TD, UK
| | - Harry Campbell
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH8 9AG, UK
| | - Tōnu Esko
- Estonian Genome Center, University of Tartu, University of Tartu, Tartu, 51010, Estonia.,Program in Medical and Population Genetics, Broad Institute, Broad Institute, Cambridge, MA, 02142, USA
| | - Zoltán Kutalik
- Institute of Social and Preventive Medicine, Lausanne University Hospital, Lausanne, 1010, Switzerland.,Swiss Institute of Bioinformatics, Lausanne, 1015, Switzerland
| | - James F Wilson
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH8 9AG, UK.,MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU, UK
| |
Collapse
|
86
|
Steinhoff G, Nesteruk J, Wolfien M, Große J, Ruch U, Vasudevan P, Müller P. Stem cells and heart disease - Brake or accelerator? Adv Drug Deliv Rev 2017; 120:2-24. [PMID: 29054357 DOI: 10.1016/j.addr.2017.10.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 10/12/2017] [Accepted: 10/13/2017] [Indexed: 12/11/2022]
Abstract
After two decades of intensive research and attempts of clinical translation, stem cell based therapies for cardiac diseases are not getting closer to clinical success. This review tries to unravel the obstacles and focuses on underlying mechanisms as the target for regenerative therapies. At present, the principal outcome in clinical therapy does not reflect experimental evidence. It seems that the scientific obstacle is a lack of integration of knowledge from tissue repair and disease mechanisms. Recent insights from clinical trials delineate mechanisms of stem cell dysfunction and gene defects in repair mechanisms as cause of atherosclerosis and heart disease. These findings require a redirection of current practice of stem cell therapy and a reset using more detailed analysis of stem cell function interfering with disease mechanisms. To accelerate scientific development the authors suggest intensifying unified computational data analysis and shared data knowledge by using open-access data platforms.
Collapse
Affiliation(s)
- Gustav Steinhoff
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Julia Nesteruk
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Markus Wolfien
- University Rostock, Institute of Computer Science, Department of Systems Biology and Bioinformatics, Ulmenstraße 69, 18057 Rostock, Germany.
| | - Jana Große
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Ulrike Ruch
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Praveen Vasudevan
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Paula Müller
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| |
Collapse
|
87
|
Mazzatenta A, Carluccio A, Robbe D, Giulio CD, Cellerino A. The companion dog as a unique translational model for aging. Semin Cell Dev Biol 2017; 70:141-153. [DOI: 10.1016/j.semcdb.2017.08.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 08/06/2017] [Accepted: 08/07/2017] [Indexed: 10/19/2022]
|
88
|
|
89
|
Tindale LC, Leach S, Spinelli JJ, Brooks-Wilson AR. Lipid and Alzheimer's disease genes associated with healthy aging and longevity in healthy oldest-old. Oncotarget 2017; 8:20612-20621. [PMID: 28206976 PMCID: PMC5400530 DOI: 10.18632/oncotarget.15296] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 01/08/2017] [Indexed: 12/20/2022] Open
Abstract
Several studies have found that long-lived individuals do not appear to carry lower numbers of common disease-associated variants than ordinary people; it has been hypothesized that they may instead carry protective variants. An intriguing type of protective variant is buffering variants that protect against variants that have deleterious effects. We genotyped 18 variants in 15 genes related to longevity or healthy aging that had been previously reported as having a gene-gene interaction or buffering effect. We compared a group of 446 healthy oldest-old ‘Super-Seniors’ (individuals 85 or older who have never been diagnosed with cancer, cardiovascular disease, dementia, diabetes or major pulmonary disease) to 421 random population-based midlife controls. Cases and controls were of European ancestry. Association tests of individual SNPs showed that Super-Seniors were less likely than controls to carry an APOEε4 allele or a haptoglobin HP2 allele. Interactions between APOE/FOXO3, APOE/CRYL1, and LPA/CRYL1 did not remain significant after multiple testing correction. In a network analysis of the candidate genes, lipid and cholesterol metabolism was a common theme. APOE, HP, and CRYL1 have all been associated with Alzheimer’s Disease, the pathology of which involves lipid and cholesterol pathways. Age-related changes in lipid and cholesterol maintenance, particularly in the brain, may be central to healthy aging and longevity.
Collapse
Affiliation(s)
- Lauren C Tindale
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, B.C., Canada.,Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, B.C., Canada
| | - Stephen Leach
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, B.C., Canada
| | - John J Spinelli
- Cancer Control Research, British Columbia Cancer Agency, Vancouver, B.C., Canada.,School of Population and Public Health, University of British Columbia, Vancouver, B.C., Canada
| | - Angela R Brooks-Wilson
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, B.C., Canada.,Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, B.C., Canada
| |
Collapse
|
90
|
Eline Slagboom P, van den Berg N, Deelen J. Phenome and genome based studies into human ageing and longevity: An overview. Biochim Biophys Acta Mol Basis Dis 2017; 1864:2742-2751. [PMID: 28951210 DOI: 10.1016/j.bbadis.2017.09.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 09/11/2017] [Accepted: 09/15/2017] [Indexed: 12/13/2022]
Abstract
Human ageing is an extremely personal process leading across the life course of individuals to large population heterogeneity in the decline of functional capacity, health and lifespan. The extremes of this process are witnessed by the healthy vital 100-year-olds on one end and the 60-year-olds suffering from multiple morbid conditions on the other end of the spectrum. Molecular studies into the basis of this heterogeneity have focused on a range of endpoints and methodological approaches. The phenotype definitions most prominently investigated in these studies are either lifespan-related or biomarker based indices of the biological ageing rate of individuals and their tissues. Unlike for many complex, age-related diseases, consensus on the ultimate set of multi-biomarker ageing or lifespan-related phenotypes for genetic and genomic studies has not been reached yet. Comparable to animal models, hallmarks of age-related disease risk, healthy ageing and longevity include immune and metabolic pathways. Potentially novel genomic regions and pathways have been identified among many (epi)genomic studies into chronological age and studies into human lifespan regulation, with APOE and FOXO3A representing yet the most robust loci. Functional analysis of a handful of genes in cell-based and animal models is ongoing. The way forward in human ageing and longevity studies seems through improvements in the interpretation of the biology of the genome, in application of computational and systems biology, integration with animal models and by harmonization of repeated phenotypic and omics measures in longitudinal and intervention studies. This article is part of a Special Issue entitled: Model Systems of Aging - edited by "Houtkooper Riekelt".
Collapse
Affiliation(s)
- P Eline Slagboom
- Department of Molecular Epidemiology, Leiden University Medical Center, Albinusdreef 2, 2333, ZA, Leiden, The Netherlands.
| | - Niels van den Berg
- Department of Molecular Epidemiology, Leiden University Medical Center, Albinusdreef 2, 2333, ZA, Leiden, The Netherlands.
| | - Joris Deelen
- Department of Molecular Epidemiology, Leiden University Medical Center, Albinusdreef 2, 2333, ZA, Leiden, The Netherlands; Max Planck Institute for Biology of Ageing; Joseph-Stelzmann-Str. 9b, D-50931 Köln (Cologne), Germany.
| |
Collapse
|
91
|
Cardiac Function Improvement and Bone Marrow Response -: Outcome Analysis of the Randomized PERFECT Phase III Clinical Trial of Intramyocardial CD133 + Application After Myocardial Infarction. EBioMedicine 2017; 22:208-224. [PMID: 28781130 PMCID: PMC5552265 DOI: 10.1016/j.ebiom.2017.07.022] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 07/18/2017] [Accepted: 07/24/2017] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE The phase III clinical trial PERFECT was designed to assess clinical safety and efficacy of intramyocardial CD133+ bone marrow stem cell treatment combined with CABG for induction of cardiac repair. DESIGN Multicentre, double-blinded, randomised placebo controlled trial. SETTING The study was conducted across six centres in Germany October 2009 through March 2016 and stopped due slow recruitment after positive interim analysis in March 2015. PARTICIPANTS Post-infarction patients with chronic ischemia and reduced LVEF (25-50%). INTERVENTIONS Eighty-two patients were randomised to two groups receiving intramyocardial application of 5ml placebo or a suspension of 0.5-5×106 CD133+. OUTCOME Primary endpoint was delta (∆) LVEF at 180days (d) compared to baseline measured in MRI. FINDINGS (PRESPECIFIED) Safety (n=77): 180d survival was 100%, MACE n=2, SAE n=49, without difference between placebo and CD133+. Efficacy (n=58): The LVEF improved from baseline LVEF 33.5% by +9.6% at 180d, p=0.001 (n=58). Treatment groups were not different in ∆LVEF (ANCOVA: Placebo +8.8% vs. CD133+ +10.4%, ∆CD133+vs placebo +2.6%, p=0.4). FINDINGS (POST HOC) Responders (R) classified by ∆LVEF≥5% after 180d were 60% of the patients (35/58) in both treatment groups. ∆LVEF in ANCOVA was +17.1% in (R) vs. non-responders (NR) (∆LVEF 0%, n=23). NR were characterized by a preoperative response signature in peripheral blood with reduced CD133+ EPC (RvsNR: p=0.005) and thrombocytes (p=0.004) in contrast to increased Erythropoeitin (p=0.02), and SH2B3 mRNA expression (p=0.073). Actuarial computed mean survival time was 76.9±3.32months (R) vs. +72.3±5.0months (NR), HR 0.3 [Cl 0.07-1.2]; p=0.067.Using a machine learning 20 biomarker response parameters were identified allowing preoperative discrimination with an accuracy of 80% (R) and 84% (NR) after 10-fold cross-validation. INTERPRETATION The PERFECT trial analysis demonstrates that the regulation of induced cardiac repair is linked to the circulating pool of CD133+ EPC and thrombocytes, associated with SH2B3 gene expression. Based on these findings, responders to cardiac functional improvement may be identified by a peripheral blood biomarker signature. TRIAL REGISTRATION ClinicalTrials.govNCT00950274.
Collapse
|
92
|
McDaid AF, Joshi PK, Porcu E, Komljenovic A, Li H, Sorrentino V, Litovchenko M, Bevers RPJ, Rüeger S, Reymond A, Bochud M, Deplancke B, Williams RW, Robinson-Rechavi M, Paccaud F, Rousson V, Auwerx J, Wilson JF, Kutalik Z. Bayesian association scan reveals loci associated with human lifespan and linked biomarkers. Nat Commun 2017; 8:15842. [PMID: 28748955 PMCID: PMC5537485 DOI: 10.1038/ncomms15842] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 05/08/2017] [Indexed: 02/07/2023] Open
Abstract
The enormous variation in human lifespan is in part due to a myriad of sequence variants, only a few of which have been revealed to date. Since many life-shortening events are related to diseases, we developed a Mendelian randomization-based method combining 58 disease-related GWA studies to derive longevity priors for all HapMap SNPs. A Bayesian association scan, informed by these priors, for parental age of death in the UK Biobank study (n=116,279) revealed 16 independent SNPs with significant Bayes factor at a 5% false discovery rate (FDR). Eleven of them replicate (5% FDR) in five independent longevity studies combined; all but three are depleted of the life-shortening alleles in older Biobank participants. Further analysis revealed that brain expression levels of nearby genes (RBM6, SULT1A1 and CHRNA5) might be causally implicated in longevity. Gene expression and caloric restriction experiments in model organisms confirm the conserved role for RBM6 and SULT1A1 in modulating lifespan.
Collapse
Affiliation(s)
- Aaron F McDaid
- Institute of Social and Preventive Medicine (IUMSP), Lausanne University Hospital, Lausanne 1010, Switzerland.,Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
| | - Peter K Joshi
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh EH8 9AG, Scotland
| | - Eleonora Porcu
- Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland.,Center for Integrative Genomics, University of Lausanne, Lausanne 1015, Switzerland
| | - Andrea Komljenovic
- Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland.,Department of Ecology and Evolution, University of Lausanne, Lausanne 1015, Switzerland
| | - Hao Li
- Laboratory of Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Vincenzo Sorrentino
- Laboratory of Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Maria Litovchenko
- Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland.,Laboratory of Systems Biology and Genetics, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne and Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
| | - Roel P J Bevers
- Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland.,Laboratory of Systems Biology and Genetics, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne and Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
| | - Sina Rüeger
- Institute of Social and Preventive Medicine (IUMSP), Lausanne University Hospital, Lausanne 1010, Switzerland.,Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
| | - Alexandre Reymond
- Center for Integrative Genomics, University of Lausanne, Lausanne 1015, Switzerland
| | - Murielle Bochud
- Institute of Social and Preventive Medicine (IUMSP), Lausanne University Hospital, Lausanne 1010, Switzerland
| | - Bart Deplancke
- Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland.,Laboratory of Systems Biology and Genetics, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne and Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
| | - Robert W Williams
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - Marc Robinson-Rechavi
- Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland.,Department of Ecology and Evolution, University of Lausanne, Lausanne 1015, Switzerland
| | - Fred Paccaud
- Institute of Social and Preventive Medicine (IUMSP), Lausanne University Hospital, Lausanne 1010, Switzerland
| | - Valentin Rousson
- Institute of Social and Preventive Medicine (IUMSP), Lausanne University Hospital, Lausanne 1010, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - James F Wilson
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh EH8 9AG, Scotland.,MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, Scotland
| | - Zoltán Kutalik
- Institute of Social and Preventive Medicine (IUMSP), Lausanne University Hospital, Lausanne 1010, Switzerland.,Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
| |
Collapse
|
93
|
Demographic, genetic and phenotypic characteristics of centenarians in Italy: Focus on gender differences. Mech Ageing Dev 2017; 165:68-74. [DOI: 10.1016/j.mad.2017.04.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 03/22/2017] [Accepted: 04/28/2017] [Indexed: 11/22/2022]
|
94
|
Dato S, Rose G, Crocco P, Monti D, Garagnani P, Franceschi C, Passarino G. The genetics of human longevity: an intricacy of genes, environment, culture and microbiome. Mech Ageing Dev 2017; 165:147-155. [DOI: 10.1016/j.mad.2017.03.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 03/04/2017] [Accepted: 03/30/2017] [Indexed: 12/13/2022]
|
95
|
Giuliani C, Pirazzini C, Delledonne M, Xumerle L, Descombes P, Marquis J, Mengozzi G, Monti D, Bellizzi D, Passarino G, Luiselli D, Franceschi C, Garagnani P. Centenarians as extreme phenotypes: An ecological perspective to get insight into the relationship between the genetics of longevity and age-associated diseases. Mech Ageing Dev 2017; 165:195-201. [DOI: 10.1016/j.mad.2017.02.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 02/14/2017] [Accepted: 02/20/2017] [Indexed: 10/20/2022]
|
96
|
Cognitive status in the oldest old and centenarians: a condition crucial for quality of life methodologically difficult to assess. Mech Ageing Dev 2017; 165:185-194. [PMID: 28286214 DOI: 10.1016/j.mad.2017.02.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 01/26/2017] [Accepted: 02/07/2017] [Indexed: 12/20/2022]
Abstract
Human life expectancy and the number of the oldest old are rapidly increasing worldwide. Advanced age is the main risk factor for dementia, representing one of the major causes of disability/dependency among older people with a strong impact on their families/caregivers. Centenarians have reached the extreme limits of human life escaping or delaying the major age-related diseases. Thus, these extraordinary individuals embody the best model to answer the crucial question if cognitive decline and dementia are progressive and unavoidable occurrences of increasing age. Despite a growing amount of data underlines the importance of cognitive function for quality of life and survival in old age, studies on centenarians have paid more attention to their physical condition rather than the assessment of their actual cognitive abilities. Accordingly, this work aims to summarize available data on the prevalence of dementia in centenarians and to critically address topics which can have a relevant impact on the cognitive assessment/status of the oldest old: (i) lack of standardized tools for cognitive assessment; (ii) criteria and threshold to establish the presence of dementia; (iii) influence of birth cohort and education; (iv) role of depression or positive attitude towards life; (v) gender differences.
Collapse
|
97
|
|
98
|
Zhang YX, Su Y, Tang L, Yang ZX, Zhou DF, Qiu YM, Cai WW. CETP polymorphisms confer genetic contribution to centenarians of Hainan, south of China. ASIAN PAC J TROP MED 2016; 9:872-876. [PMID: 27633301 DOI: 10.1016/j.apjtm.2016.07.009] [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: 05/17/2016] [Revised: 06/16/2016] [Accepted: 07/01/2016] [Indexed: 10/21/2022] Open
Abstract
OBJECTIVE In this paper, we will discuss if the CETP polymorphism contributes to the centenarians in Hainan island. METHODS We tested the TaqIB and I405V polymorphisms of CETP gene among 276 centenarians and 301 matched healthy individuals by AS-PCR and analyzed the data with SPSS software package (Version 19.0). RESULTS Our data indicated that allele B1 and V have the significant differences between centenarians and healthy control groups with P < 0.001. Further analysis implied that genotypes B1B1 (P < 0.001, OR = 0.148, 95% CI = 0.095-0.230) and VV (P < 0.001 and OR = 0.353, 95% CI = 0.237-0.525) were significantly different between centenarians and matched controls. The combination of B and V, such as B1B1-II (P < 0.001, OR = 0.128, 95% CI = 0.049-0.329), B1B1-IV (P < 0.001, OR = 0.115, 95% CI = 0.056-0.237), B1B2-VV (P < 0.05, OR = 0.534, 95% CI = 0.310-0.920), and B2B2-VV (P < 0.001, OR = 0.198, 95% CI = 0.086-0.453) have significant differences between centenarians and matched healthy individuals from Hainan. CONCLUSION Our results implied that allele B1B1 and VV, as well as the combination B1B1-II, B1B1-IV, B1B2-VV and B2B2-VV may contribute to the longevity in centenarians of Hainan, south of China.
Collapse
Affiliation(s)
- Yun-Xia Zhang
- Department of Biochemistry and Molecular Biology, Hainan Medical College, Haikou, Hainan Province, China
| | - Ya Su
- Hainan Cadre Sanatorium/Geriatric Hospital of Hainan, Haikou, Hainan Province, China
| | - Li Tang
- Department of Reproduction and Genetics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Ze-Xing Yang
- Department of Reproduction and Genetics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Dai-Feng Zhou
- Department of Biochemistry and Molecular Biology, Hainan Medical College, Haikou, Hainan Province, China
| | - Yi-Min Qiu
- Department of Biochemistry and Molecular Biology, Hainan Medical College, Haikou, Hainan Province, China
| | - Wang-Wei Cai
- Department of Biochemistry and Molecular Biology, Hainan Medical College, Haikou, Hainan Province, China.
| |
Collapse
|
99
|
Arey RN, Murphy CT. Conserved regulators of cognitive aging: From worms to humans. Behav Brain Res 2016; 322:299-310. [PMID: 27329151 DOI: 10.1016/j.bbr.2016.06.035] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 05/27/2016] [Accepted: 06/17/2016] [Indexed: 01/25/2023]
Abstract
Cognitive decline is a major deficit that arises with age in humans. While some research on the underlying causes of these problems can be done in humans, harnessing the strengths of small model systems, particularly those with well-studied longevity mutants, such as the nematode C. elegans, will accelerate progress. Here we review the approaches being used to study cognitive decline in model organisms and show how simple model systems allow the rapid discovery of conserved molecular mechanisms, which will eventually enable the development of therapeutics to slow cognitive aging.
Collapse
Affiliation(s)
- Rachel N Arey
- Department of Molecular Biology & LSI Genomics, Princeton University, Princeton, NJ 08544, United States
| | - Coleen T Murphy
- Department of Molecular Biology & LSI Genomics, Princeton University, Princeton, NJ 08544, United States.
| |
Collapse
|
100
|
Erikson GA, Bodian DL, Rueda M, Molparia B, Scott ER, Scott-Van Zeeland AA, Topol SE, Wineinger NE, Niederhuber JE, Topol EJ, Torkamani A. Whole-Genome Sequencing of a Healthy Aging Cohort. Cell 2016; 165:1002-11. [PMID: 27114037 DOI: 10.1016/j.cell.2016.03.022] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 02/05/2016] [Accepted: 03/14/2016] [Indexed: 01/23/2023]
Abstract
Studies of long-lived individuals have revealed few genetic mechanisms for protection against age-associated disease. Therefore, we pursued genome sequencing of a related phenotype-healthy aging-to understand the genetics of disease-free aging without medical intervention. In contrast with studies of exceptional longevity, usually focused on centenarians, healthy aging is not associated with known longevity variants, but is associated with reduced genetic susceptibility to Alzheimer and coronary artery disease. Additionally, healthy aging is not associated with a decreased rate of rare pathogenic variants, potentially indicating the presence of disease-resistance factors. In keeping with this possibility, we identify suggestive common and rare variant genetic associations implying that protection against cognitive decline is a genetic component of healthy aging. These findings, based on a relatively small cohort, require independent replication. Overall, our results suggest healthy aging is an overlapping but distinct phenotype from exceptional longevity that may be enriched with disease-protective genetic factors. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Galina A Erikson
- The Scripps Translational Science Institute, Scripps Health, and The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Dale L Bodian
- Inova Translational Medicine Institute, Inova Health System, Falls Church, VA 22042 USA
| | - Manuel Rueda
- The Scripps Translational Science Institute, Scripps Health, and The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Bhuvan Molparia
- The Scripps Translational Science Institute, Scripps Health, and The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Erick R Scott
- The Scripps Translational Science Institute, Scripps Health, and The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | - Sarah E Topol
- The Scripps Translational Science Institute, Scripps Health, and The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Nathan E Wineinger
- The Scripps Translational Science Institute, Scripps Health, and The Scripps Research Institute, La Jolla, CA 92037, USA
| | - John E Niederhuber
- Inova Translational Medicine Institute, Inova Health System, Falls Church, VA 22042 USA; Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Eric J Topol
- The Scripps Translational Science Institute, Scripps Health, and The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Ali Torkamani
- The Scripps Translational Science Institute, Scripps Health, and The Scripps Research Institute, La Jolla, CA 92037, USA; Cypher Genomics, Inc., San Diego, CA 92121, USA.
| |
Collapse
|