1
|
Ying K, Castro JP, Shindyapina AV, Tyshkovskiy A, Moqri M, Goeminne LJE, Milman S, Zhang ZD, Barzilai N, Gladyshev VN. Depletion of loss-of-function germline mutations in centenarians reveals longevity genes. Nat Commun 2024; 15:9030. [PMID: 39424787 PMCID: PMC11489729 DOI: 10.1038/s41467-024-52967-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 09/27/2024] [Indexed: 10/21/2024] Open
Abstract
While previous studies identified common genetic variants associated with longevity in centenarians, the role of the rare loss-of-function (LOF) mutation burden remains largely unexplored. Here, we investigated the burden of rare LOF mutations in Ashkenazi Jewish individuals from the Longevity Genes Project and LonGenity study cohorts using whole-exome sequencing data. We found that centenarians had a significantly lower burden (11-22%) of LOF mutations compared to controls. Similar effects were also observed in their offspring. Gene-level burden analysis identified 35 genes with depleted LOF mutations in centenarians, with 14 of these validated in the UK Biobank. Mendelian randomization and multi-omic analyses on these genes identified RGP1, PCNX2, and ANO9 as longevity genes with consistent causal effects on multiple aging-related traits and altered expression during aging. Our findings suggest that a protective genetic background, characterized by a reduced burden of damaging variants, contributes to exceptional longevity, likely acting in concert with specific protective variants to promote healthy aging.
Collapse
Affiliation(s)
- Kejun Ying
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, USA
- T. H. Chan School of Public Health, Harvard University, Boston, USA
| | - José P Castro
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, USA
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto and Aging and Aneuploidy Laboratory, IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Anastasia V Shindyapina
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, USA
- Retro Biosciences, Redwood City, USA
| | - Alexander Tyshkovskiy
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, USA
| | - Mahdi Moqri
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, USA
| | - Ludger J E Goeminne
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, USA
| | - Sofiya Milman
- Department of Genetics, Albert Einstein College of Medicine, Bronx, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, USA
| | - Zhengdong D Zhang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, USA
| | - Nir Barzilai
- Department of Genetics, Albert Einstein College of Medicine, Bronx, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, USA
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, USA.
| |
Collapse
|
2
|
Zhang Y, Murata S, Schmidt-Mende K, Ebeling M, Modig K. Do people reach 100 by surviving, delaying, or avoiding diseases? A life course comparison of centenarians and non-centenarians from the same birth cohorts. GeroScience 2024:10.1007/s11357-024-01330-w. [PMID: 39212787 DOI: 10.1007/s11357-024-01330-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
Centenarians are perceived as pioneers of longevity, possessing the secrets to surpassing age 100. It remains unclear whether they achieve this by surviving, delaying, or avoiding diseases to a greater extent than their shorter-lived peers. This register-based study encompassed all individuals aged 60 and older, born between 1912 and 1922 in Stockholm County, Sweden (N = 170,787). Using historical data, individuals were prospectively followed from 1972 to 2022 and stratified by their age at death. Age-specific incidence rates and remaining lifetime risk from age 60 were calculated for stroke, myocardial infarction, hip fracture, and various cancers (including colorectal, breast, and prostate), and compared between those who survived to age 100 and their shorter-lived counterparts. Centenarians had lower age-specific incidence rates for almost all diseases and ages. Despite longer life spans, their lifetime risks for all diseases except hip fracture were lower than those of non-centenarians. This suggests that centenarians delay, and even avoid, many of the major age-related diseases rather than surviving them to a higher extent. The findings that centenarians not only exhibit lower disease rates at younger ages compared to their shorter-lived peers but throughout their lives challenge the notion that longer life span inevitably leads to higher disease rates or a simple shift of diseases to older ages.
Collapse
Affiliation(s)
- Yuge Zhang
- Unit of Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Box 210, 17177, Stockholm, Sweden.
| | - Shunsuke Murata
- Unit of Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Box 210, 17177, Stockholm, Sweden
| | - Katharina Schmidt-Mende
- Academic Primary Health Care Centre, Stockholm Region, Stockholm, Sweden
- Division of Family Medicine and Primary Care, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
| | - Marcus Ebeling
- Unit of Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Box 210, 17177, Stockholm, Sweden
- Max Planck Institute for Demographic Research, Rostock, Germany
| | - Karin Modig
- Unit of Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Box 210, 17177, Stockholm, Sweden
| |
Collapse
|
3
|
Smulders L, Deelen J. Genetics of human longevity: From variants to genes to pathways. J Intern Med 2024; 295:416-435. [PMID: 37941149 DOI: 10.1111/joim.13740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Abstract
The current increase in lifespan without an equivalent increase in healthspan poses a grave challenge to the healthcare system and a severe burden on society. However, some individuals seem to be able to live a long and healthy life without the occurrence of major debilitating chronic diseases, and part of this trait seems to be hidden in their genome. In this review, we discuss the findings from studies on the genetic component of human longevity and the main challenges accompanying these studies. We subsequently focus on results from genetic studies in model organisms and comparative genomic approaches to highlight the most important conserved longevity-associated pathways. By combining the results from studies using these different approaches, we conclude that only five main pathways have been consistently linked to longevity, namely (1) insulin/insulin-like growth factor 1 signalling, (2) DNA-damage response and repair, (3) immune function, (4) cholesterol metabolism and (5) telomere maintenance. As our current approaches to study the relevance of these pathways in humans are limited, we suggest that future studies on the genetics of human longevity should focus on the identification and functional characterization of rare genetic variants in genes involved in these pathways.
Collapse
Affiliation(s)
- Larissa Smulders
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Joris Deelen
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| |
Collapse
|
4
|
Maksiutenko EM, Merkureva V, Barbitoff YA, Tsay VV, Aseev MV, Glotov AS, Glotov OS. Exome sequencing in extreme altitude mountaineers identifies pathogenic variants in RTEL1 and COL6A1 previously associated with respiratory failure. Physiol Rep 2024; 12:e16015. [PMID: 38653581 PMCID: PMC11039409 DOI: 10.14814/phy2.16015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 04/02/2024] [Accepted: 04/02/2024] [Indexed: 04/25/2024] Open
Abstract
Adaptation of humans to challenging environmental conditions, such as extreme temperature, malnutrition, or hypoxia, is an interesting phenomenon for both basic and applied research. Identification of the genetic factors contributing to human adaptation to these conditions enhances our understanding of the underlying molecular and physiological mechanisms. In our study, we analyzed the exomes of 22 high altitude mountaineers to uncover genetic variants contributing to hypoxic adaptation. To our surprise, we identified two putative loss-of-function variants, rs1385101139 in RTEL1 and rs1002726737 in COL6A1 in two extremely high altitude (personal record of more than 8500 m) professional climbers. Both variants can be interpreted as pathogenic according to medical geneticists' guidelines, and are linked to inherited conditions involving respiratory failure (late-onset pulmonary fibrosis and severe Ullrich muscular dystrophy for rs1385101139 and rs1002726737, respectively). Our results suggest that a loss of gene function may act as an important factor of human adaptation, which is corroborated by previous reports in other human subjects.
Collapse
Affiliation(s)
- Evgeniia M. Maksiutenko
- Department of Genomic MedicineD.O. Ott Research Institute of Obstetrics, Gynaecology, and ReproductologySt. PetersburgRussia
| | - Valeriia Merkureva
- Department of Genomic MedicineD.O. Ott Research Institute of Obstetrics, Gynaecology, and ReproductologySt. PetersburgRussia
- CerbaLab Ltd.St. PetersburgRussia
| | - Yury A. Barbitoff
- Department of Genomic MedicineD.O. Ott Research Institute of Obstetrics, Gynaecology, and ReproductologySt. PetersburgRussia
- Department of Genetics and BiotechnologySt. Petersburg State UniversitySt. PetersburgRussia
| | - Victoria V. Tsay
- Department of Genomic MedicineD.O. Ott Research Institute of Obstetrics, Gynaecology, and ReproductologySt. PetersburgRussia
- CerbaLab Ltd.St. PetersburgRussia
- Department of Experimental Medical VirologyMolecular Genetics and Biobanking of Pediatric Research and Clinical Center for Infectious DiseasesSt. PetersburgRussia
| | - Mikhail V. Aseev
- Department of Genomic MedicineD.O. Ott Research Institute of Obstetrics, Gynaecology, and ReproductologySt. PetersburgRussia
- CerbaLab Ltd.St. PetersburgRussia
| | - Andrey S. Glotov
- Department of Genomic MedicineD.O. Ott Research Institute of Obstetrics, Gynaecology, and ReproductologySt. PetersburgRussia
- Department of Genetics and BiotechnologySt. Petersburg State UniversitySt. PetersburgRussia
| | - Oleg S. Glotov
- Department of Genomic MedicineD.O. Ott Research Institute of Obstetrics, Gynaecology, and ReproductologySt. PetersburgRussia
- Department of Experimental Medical VirologyMolecular Genetics and Biobanking of Pediatric Research and Clinical Center for Infectious DiseasesSt. PetersburgRussia
| |
Collapse
|
5
|
Milman S, Barzilai N. Discovering Biological Mechanisms of Exceptional Human Health Span and Life Span. Cold Spring Harb Perspect Med 2023; 13:a041204. [PMID: 37137499 PMCID: PMC10513160 DOI: 10.1101/cshperspect.a041204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Humans age at different rates and families with exceptional longevity provide an opportunity to understand why some people age slower than others. Unique features exhibited by centenarians include a family history of extended life span, compression of morbidity with resultant extension of health span, and longevity-associated biomarker profiles. These biomarkers, including low-circulating insulin-like growth factor 1 (IGF-1) and elevated high-density lipoprotein (HDL) cholesterol levels, are associated with functional genotypes that are enriched in centenarians, suggesting that they may be causative for longevity. While not all genetic discoveries from centenarians have been validated, in part due to exceptional life span being a rare phenotype in the general population, the APOE2 and FOXO3a genotypes have been confirmed in a number of populations with exceptional longevity. However, life span is now recognized as a complex trait and genetic research methods to study longevity are rapidly extending beyond classical Mendelian genetics to polygenic inheritance methodologies. Moreover, newer approaches are suggesting that pathways that have been recognized for decades to control life span in animals may also regulate life span in humans. These discoveries led to strategic development of therapeutics that may delay aging and prolong health span.
Collapse
Affiliation(s)
- Sofiya Milman
- Institute for Aging Research, Department of Medicine, Divisions of Endocrinology and Geriatrics, Department of Genetics, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Nir Barzilai
- Institute for Aging Research, Department of Medicine, Divisions of Endocrinology and Geriatrics, Department of Genetics, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| |
Collapse
|
6
|
Trofimiuk-Müldner M, Domagała B, Sokołowski G, Skalniak A, Hubalewska-Dydejczyk A. AIP gene germline variants in adult Polish patients with apparently sporadic pituitary macroadenomas. Front Endocrinol (Lausanne) 2023; 14:1098367. [PMID: 36843582 PMCID: PMC9950257 DOI: 10.3389/fendo.2023.1098367] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/23/2023] [Indexed: 02/12/2023] Open
Abstract
INTRODUCTION Up to 5% of all pituitary tumors are hereditary e.g. due to MEN1 or aryl hydrocarbon receptor-interacting protein (AIP) genes mutations. OBJECTIVES The study was aimed at the assessment of the frequency and characteristics of AIP-mutation related tumors in patients with apparently sporadic pituitary macroadenomas in the Polish population. MATERIALS AND METHODS The study included 131 patients (57 males, 74 females; median age 42 years) diagnosed with pituitary macroadenomas, and with a negative family history of familial isolated pituitary adenoma (FIPA) or multiple endocrine neoplasia type 1 (MEN1) syndromes. Sanger sequencing was used for the assessment of AIP gene variants. The study was approved by the Ethics Board of JUMC. RESULTS AIP variants were identified in five of the 131 included subjects (3.8%): one diagnosed with Cushing's disease, two with acromegaly, and two with non-secreting adenomas. Patients harboring hereditary AIP gene alterations did not differ from the rest of the study group in median age at diagnosis (41.0 vs. 42.5 years, P=0.8), median largest tumor diameter (25 vs. 24 mm, P=0.6), gender distribution (60.0% vs. 56.3% females, P=0.8), secreting tumor frequency (60.0% vs. 67.5%, P=0.7), or acromegaly diagnosis frequency (40.0% vs.37.3%, P=0.9). CONCLUSIONS In our series of apparently sporadic pituitary macroadenomas, AIP gene variant carriers did not differ substantially from patients with negative genetic testing. A risk factor-centred approach to AIP genetic screening may result in missing germline variants. Considering the clinical impact of such genetic variants and their relatively low penetrance, it is, however, doubtful if general genetic screening benefits the whole cohort of pituitary macroadenoma patients and their families.
Collapse
Affiliation(s)
- Małgorzata Trofimiuk-Müldner
- Chair and Department of Endocrinology, Jagiellonian University Medical College, Kraków, Poland
- *Correspondence: Małgorzata Trofimiuk-Müldner,
| | - Bartosz Domagała
- Department of Endocrinology, Endocrine Oncology and Nuclear Medicine, University Hospital in Kraków, Kraków, Poland
| | - Grzegorz Sokołowski
- Chair and Department of Endocrinology, Jagiellonian University Medical College, Kraków, Poland
| | - Anna Skalniak
- Chair and Department of Endocrinology, Jagiellonian University Medical College, Kraków, Poland
| | | |
Collapse
|
7
|
Nazarian A, Loika Y, He L, Culminskaya I, Kulminski AM. Genome-wide analysis identified abundant genetic modulators of contributions of the apolipoprotein E alleles to Alzheimer's disease risk. Alzheimers Dement 2022; 18:2067-2078. [PMID: 34978151 PMCID: PMC9250541 DOI: 10.1002/alz.12540] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 08/31/2021] [Accepted: 10/25/2021] [Indexed: 01/31/2023]
Abstract
INTRODUCTION The apolipoprotein E (APOE) ε2 and ε4 alleles have beneficial and adverse impacts on Alzheimer's disease (AD), respectively, with incomplete penetrance, which may be modulated by other genetic variants. METHODS We examined whether the associations of the APOE alleles with other polymorphisms in the genome can be sensitive to AD-affection status. RESULTS We identified associations of the ε2 and ε4 alleles with 314 and 232 polymorphisms, respectively. Of them, 35 and 31 polymorphisms had significantly different effects in AD-affected and -unaffected groups, suggesting their potential involvement in the AD pathogenesis by modulating the effects of the ε2 and ε4 alleles, respectively. Our survival-type analysis of the AD risk supported modulating roles of multiple group-specific polymorphisms. Our functional analysis identified gene enrichment in multiple immune-related biological processes, for example, B cell function. DISCUSSION These findings suggest involvement of local and inter-chromosomal modulators of the effects of the APOE alleles on the AD risk.
Collapse
Affiliation(s)
- Alireza Nazarian
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, USA
| | - Yury Loika
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, USA
| | - Liang He
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, USA
| | - Irina Culminskaya
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, USA
| | - Alexander M. Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, USA
| |
Collapse
|
8
|
Shchagina O, Fedotov V, Markova T, Shatokhina O, Ryzhkova O, Fedotova T, Polyakov A. Palmoplantar Keratoderma: A Molecular Genetic Analysis of Family Cases. Int J Mol Sci 2022; 23:ijms23179576. [PMID: 36076978 PMCID: PMC9455982 DOI: 10.3390/ijms23179576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/14/2022] [Accepted: 08/17/2022] [Indexed: 11/16/2022] Open
Abstract
Palmoplantar keratoderma is a clinically polymorphic disorder with a heterogeneous etiology characterized by marked hyperkeratotic lesions on the surface of palms and soles. Hereditary forms of palmoplantar keratoderma usually have autosomal dominant inheritance and are caused by mutations in dozens of genes, most of which belong to the keratin family. We carried out clinical and molecular genetic analysis of the affected and healthy members of four families with autosomal dominant palmoplantar keratoderma. In three out of four family cases of autosomal dominant palmoplantar keratoderma, the following molecular genetic causes were established: in two families—previously non-described missense mutations in the AQP5 gene (NM_001651.4): c.369C>G (p.(Asn123Lys)) and c.103T>G (p.(Trp35Gly)); in one family—a described splice site mutation in the KRT9 gene (NM_000226.4): c.31T>G. In one family, the possible cause of palmoplantar keratoderma was detected—a variant in the KRT1 gene (NM_006121.4): c.931G>A (p.(Glu311Lys)).
Collapse
Affiliation(s)
- Olga Shchagina
- Research Centre for Medical Genetics, Moskvorechye St., 1, 115522 Moscow, Russia
- Correspondence:
| | - Valeriy Fedotov
- Voronezh Regional Clinical Hospital №1, Moscow Avenue, 151, 394066 Voronezh, Russia
| | - Tatiana Markova
- Research Centre for Medical Genetics, Moskvorechye St., 1, 115522 Moscow, Russia
| | - Olga Shatokhina
- Research Centre for Medical Genetics, Moskvorechye St., 1, 115522 Moscow, Russia
| | - Oksana Ryzhkova
- Research Centre for Medical Genetics, Moskvorechye St., 1, 115522 Moscow, Russia
| | - Tatiana Fedotova
- Voronezh Regional Clinical Hospital №1, Moscow Avenue, 151, 394066 Voronezh, Russia
| | - Aleksander Polyakov
- Research Centre for Medical Genetics, Moskvorechye St., 1, 115522 Moscow, Russia
| |
Collapse
|
9
|
Nazarian A, Philipp I, Culminskaya I, He L, Kulminski AM. Inter- and intra-chromosomal modulators of the APOE ɛ2 and ɛ4 effects on the Alzheimer's disease risk. GeroScience 2022; 45:233-247. [PMID: 35809216 PMCID: PMC9886755 DOI: 10.1007/s11357-022-00617-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 06/24/2022] [Indexed: 02/03/2023] Open
Abstract
The mechanisms of incomplete penetrance of risk-modifying impacts of apolipoprotein E (APOE) ε2 and ε4 alleles on Alzheimer's disease (AD) have not been fully understood. We performed genome-wide analysis of differences in linkage disequilibrium (LD) patterns between 6,136 AD-affected and 10,555 AD-unaffected subjects from five independent studies to explore whether the association of the APOE ε2 allele (encoded by rs7412 polymorphism) and ε4 allele (encoded by rs429358 polymorphism) with AD was modulated by autosomal polymorphisms. The LD analysis identified 24 (mostly inter-chromosomal) and 57 (primarily intra-chromosomal) autosomal polymorphisms with significant differences in LD with either rs7412 or rs429358, respectively, between AD-affected and AD-unaffected subjects, indicating their potential modulatory roles. Our Cox regression analysis showed that minor alleles of four inter-chromosomal and ten intra-chromosomal polymorphisms exerted significant modulating effects on the ε2- and ε4-associated AD risks, respectively, and identified ε2-independent (rs2884183 polymorphism, 11q22.3) and ε4-independent (rs483082 polymorphism, 19q13.32) associations with AD. Our functional analysis highlighted ε2- and/or ε4-linked processes affecting the lipid and lipoprotein metabolism and cell junction organization which may contribute to AD pathogenesis. These findings provide insights into the ε2- and ε4-associated mechanisms of AD pathogenesis, underlying their incomplete penetrance.
Collapse
Affiliation(s)
- Alireza Nazarian
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Erwin Mill Building, 2024 W. Main St, Durham, NC, 27705, USA.
| | - Ian Philipp
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Erwin Mill Building, 2024 W. Main St, Durham, NC 27705 USA
| | - Irina Culminskaya
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Erwin Mill Building, 2024 W. Main St, Durham, NC 27705 USA
| | - Liang He
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Erwin Mill Building, 2024 W. Main St, Durham, NC 27705 USA
| | - Alexander M. Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Erwin Mill Building, 2024 W. Main St, Durham, NC 27705 USA
| |
Collapse
|
10
|
Gunn S, Wainberg M, Song Z, Andersen S, Boudreau R, Feitosa MF, Tan Q, Montasser ME, O'Connell JR, Stitziel N, Price N, Perls T, Schork NJ, Sebastiani P. Distribution of 54 polygenic risk scores for common diseases in long lived individuals and their offspring. GeroScience 2022; 44:719-729. [PMID: 35119614 PMCID: PMC9135909 DOI: 10.1007/s11357-022-00518-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 01/15/2022] [Indexed: 12/01/2022] Open
Abstract
A surprising and well-replicated result in genetic studies of human longevity is that centenarians appear to carry disease-associated variants in numbers similar to the general population. With the proliferation of large genome-wide association studies (GWAS) in recent years, investigators have turned to polygenic scores to leverage GWAS results into a measure of genetic risk that can better predict the risk of disease than individual significant variants alone. We selected 54 polygenic risk scores (PRSs) developed for a variety of outcomes, and we calculated their values in individuals from the New England Centenarian Study (NECS, N = 4886) and the Long Life Family Study (LLFS, N = 4577). We compared the distribution of these PRSs among exceptionally long-lived individuals (ELLI), their offspring, and controls, and we also examined their predictive values, using t-tests and regression models adjusting for sex and principal components reflecting the ancestral background of the individuals (PCs). In our analyses, we controlled for multiple testing using a Bonferroni-adjusted threshold for 54 traits. We found that only 4 of the 54 PRSs differed between ELLIs and controls in both cohorts. ELLIs had significantly lower mean PRSs for Alzheimer's disease (AD) and coronary artery disease (CAD) than controls, suggesting a genetic predisposition to extreme longevity may be mediated by reduced susceptibility to these traits. ELLIs also had significantly higher mean PRSs for improved cognitive function and parental extreme longevity. In addition, the PRS for AD was associated with a higher risk of dementia among controls but not ELLIs (p = 0.003, 0.3 in NECS, p = 0.03, 0.9 in LLFS, respectively). ELLIs have a similar burden of genetic disease risk as the general population for most traits but have a significantly lower genetic risk of AD and CAD. The lack of association between AD PRS and dementia among ELLIs suggests that the genetic risk for AD that they do have is somehow counteracted by protective genetic or environmental factors.
Collapse
Affiliation(s)
- Sophia Gunn
- Department of Biostatistics, Boston University, Crosstown Building, 801 Massachusetts Avenue 3rd Floor, Boston, MA, 02118, USA.
| | - Michael Wainberg
- Institute for Systems Biology, 401 Terry Avenue North, Seattle, WA, 98109-5263, USA
| | - Zeyuan Song
- Department of Biostatistics, Boston University, Crosstown Building, 801 Massachusetts Avenue 3rd Floor, Boston, MA, 02118, USA
| | - Stacy Andersen
- Department of Medicine, Boston University, Crosstown Building, 801 Massachusetts Avenue 3rd Floor, Boston, MA, 02118, USA
| | - Robert Boudreau
- University of Pittsburgh, 127 N. Bellefield Avenue Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Mary F Feitosa
- Department of Genetics, Washington University School of Medicine in St Louis, 660 S. Euclid Ave, St. Louis, MO, 63110-1010, USA
| | - Qihua Tan
- University of Southern Denmark, Campusvej 55, Odense M, Denmark
| | - May E Montasser
- Department of Medicine, Division of Endocrinology, Diabetes and Nutrition and Program for Personalized and Genomic Medicine, School of Medicine, University of Maryland, 620 W Lexington St, BaltimoreBaltimore, MD, 21201, USA
| | - Jeffrey R O'Connell
- Department of Medicine, Division of Endocrinology, Diabetes and Nutrition and Program for Personalized and Genomic Medicine, School of Medicine, University of Maryland, 620 W Lexington St, BaltimoreBaltimore, MD, 21201, USA
| | - Nathan Stitziel
- Department of Genetics, Washington University School of Medicine in St Louis, 660 S. Euclid Ave, St. Louis, MO, 63110-1010, USA
| | - Nathan Price
- Institute for Systems Biology, 401 Terry Avenue North, Seattle, WA, 98109-5263, USA
| | - Thomas Perls
- Department of Medicine, Boston University, Crosstown Building, 801 Massachusetts Avenue 3rd Floor, Boston, MA, 02118, USA
| | | | - Paola Sebastiani
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA, USA
| |
Collapse
|
11
|
Abstract
Junctophilins (JPHs) comprise a family of structural proteins that connect the plasma membrane to intracellular organelles such as the endo/sarcoplasmic reticulum. Tethering of these membrane structures results in the formation of highly organized subcellular junctions that play important signaling roles in all excitable cell types. There are four JPH isoforms, expressed primarily in muscle and neuronal cell types. Each JPH protein consists of 6 'membrane occupation and recognition nexus' (MORN) motifs, a joining region connecting these to another set of 2 MORN motifs, a putative alpha-helical region, a divergent region exhibiting low homology between JPH isoforms, and a carboxy-terminal transmembrane region anchoring into the ER/SR membrane. JPH isoforms play essential roles in developing and maintaining subcellular membrane junctions. Conversely, inherited mutations in JPH2 cause hypertrophic or dilated cardiomyopathy, while trinucleotide expansions in the JPH3 gene cause Huntington Disease-Like 2. Loss of JPH1 protein levels can cause skeletal myopathy, while loss of cardiac JPH2 levels causes heart failure and atrial fibrillation, among other disease. This review will provide a comprehensive overview of the JPH gene family, phylogeny, and evolutionary analysis of JPH genes and other MORN domain proteins. JPH biogenesis, membrane tethering, and binding partners will be discussed, as well as functional roles of JPH isoforms in excitable cells. Finally, potential roles of JPH isoform deficits in human disease pathogenesis will be reviewed.
Collapse
Affiliation(s)
- Stephan E Lehnart
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center Göttingen, University Medical Center Göttingen, Department of Cardiology and Pneumology, Georg-August University Göttingen, Göttingen, Germany.,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Germany
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas, United States; Departments of Molecular Physiology and Biophysics, Medicine (Cardiology), Pediatrics (Cardiology), Neuroscience, and Center for Space Medicine, Baylor College of Medicine, Houston, Texas, United States
| |
Collapse
|
12
|
Bacalini MG, Palombo F, Garagnani P, Giuliani C, Fiorini C, Caporali L, Stanzani Maserati M, Capellari S, Romagnoli M, De Fanti S, Benussi L, Binetti G, Ghidoni R, Galimberti D, Scarpini E, Arcaro M, Bonanni E, Siciliano G, Maestri M, Guarnieri B, Martucci M, Monti D, Carelli V, Franceschi C, La Morgia C, Santoro A. Association of rs3027178 polymorphism in the circadian clock gene PER1 with susceptibility to Alzheimer's disease and longevity in an Italian population. GeroScience 2021; 44:881-896. [PMID: 34921659 PMCID: PMC9135916 DOI: 10.1007/s11357-021-00477-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/15/2021] [Indexed: 12/11/2022] Open
Abstract
Many physiological processes in the human body follow a 24-h circadian rhythm controlled by the circadian clock system. Light, sensed by retina, is the predominant “zeitgeber” able to synchronize the circadian rhythms to the light-dark cycles. Circadian rhythm dysfunction and sleep disorders have been associated with aging and neurodegenerative diseases including mild cognitive impairment (MCI) and Alzheimer’s disease (AD). In the present study, we aimed at investigating the genetic variability of clock genes in AD patients compared to healthy controls from Italy. We also included a group of Italian centenarians, considered as super-controls in association studies given their extreme phenotype of successful aging. We analyzed the exon sequences of eighty-four genes related to circadian rhythms, and the most significant variants identified in this first discovery phase were further assessed in a larger independent cohort of AD patients by matrix assisted laser desorption/ionization-time of flight mass spectrometry. The results identified a significant association between the rs3027178 polymorphism in the PER1 circadian gene with AD, the G allele being protective for AD. Interestingly, rs3027178 showed similar genotypic frequencies among AD patients and centenarians. These results collectively underline the relevance of circadian dysfunction in the predisposition to AD and contribute to the discussion on the role of the relationship between the genetics of age-related diseases and of longevity.
Collapse
Affiliation(s)
- Maria Giulia Bacalini
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Laboratorio Brain Aging, Bologna, Italy
| | - Flavia Palombo
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Programma di Neurogenetica, Bologna, Italy
| | - Paolo Garagnani
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.,Applied Biomedical Research Center (CRBA), S. Orsola-Malpighi Polyclinic, Bologna, Italy.,CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", Unit of Bologna, Bologna, Italy.,Department of Laboratory Medicine, Clinical Chemistry, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.,Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate), University of Bologna, Bologna, Italy
| | - Cristina Giuliani
- Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate), University of Bologna, Bologna, Italy.,Laboratory of Molecular Anthropology and Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Claudio Fiorini
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Programma di Neurogenetica, Bologna, Italy
| | - Leonardo Caporali
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Programma di Neurogenetica, Bologna, Italy
| | | | - Sabina Capellari
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Martina Romagnoli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Programma di Neurogenetica, Bologna, Italy
| | - Sara De Fanti
- Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate), University of Bologna, Bologna, Italy.,Laboratory of Molecular Anthropology and Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Luisa Benussi
- IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Giuliano Binetti
- IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Roberta Ghidoni
- IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Daniela Galimberti
- Fondazione IRCCS Ca' Granda, Ospedale Policlinico, Milan, Italy.,Dino Ferrari Center, University of Milan, Milan, Italy
| | - Elio Scarpini
- Fondazione IRCCS Ca' Granda, Ospedale Policlinico, Milan, Italy.,Dino Ferrari Center, University of Milan, Milan, Italy
| | - Marina Arcaro
- Fondazione IRCCS Ca' Granda, Ospedale Policlinico, Milan, Italy
| | - Enrica Bonanni
- Neurology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gabriele Siciliano
- Neurology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Michelangelo Maestri
- Neurology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Biancamaria Guarnieri
- Center of Sleep Medicine, Villa Serena Hospital and Villaserena Foundation for the Research, Città S. Angelo, Pescara, Italy
| | | | - Morena Martucci
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Daniela Monti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Valerio Carelli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Programma di Neurogenetica, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Claudio Franceschi
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.,Department of Applied Mathematics, Institute of Information Technology, Mathematics and Mechanics (ITMM), Lobachevsky State University of Nizhny Novgorod-National Research University (UNN), Nizhny Novgorod, Russia
| | - Chiara La Morgia
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Programma di Neurogenetica, Bologna, Italy.,IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
| | - Aurelia Santoro
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy. .,Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate), University of Bologna, Bologna, Italy.
| |
Collapse
|
13
|
Treaster S, Karasik D, Harris MP. Footprints in the Sand: Deep Taxonomic Comparisons in Vertebrate Genomics to Unveil the Genetic Programs of Human Longevity. Front Genet 2021; 12:678073. [PMID: 34163529 PMCID: PMC8215702 DOI: 10.3389/fgene.2021.678073] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/12/2021] [Indexed: 01/09/2023] Open
Abstract
With the modern quality, quantity, and availability of genomic sequencing across species, as well as across the expanse of human populations, we can screen for shared signatures underlying longevity and lifespan. Knowledge of these mechanisms would be medically invaluable in combating aging and age-related diseases. The diversity of longevities across vertebrates is an opportunity to look for patterns of genetic variation that may signal how this life history property is regulated, and ultimately how it can be modulated. Variation in human longevity provides a unique window to look for cases of extreme lifespan within a population, as well as associations across populations for factors that influence capacity to live longer. Current large cohort studies support the use of population level analyses to identify key factors associating with human lifespan. These studies are powerful in concept, but have demonstrated limited ability to resolve signals from background variation. In parallel, the expanding catalog of sequencing and annotation from diverse species, some of which have evolved longevities well past a human lifespan, provides independent cases to look at the genomic signatures of longevity. Recent comparative genomic work has shown promise in finding shared mechanisms associating with longevity among distantly related vertebrate groups. Given the genetic constraints between vertebrates, we posit that a combination of approaches, of parallel meta-analysis of human longevity along with refined analysis of other vertebrate clades having exceptional longevity, will aid in resolving key regulators of enhanced lifespan that have proven to be elusive when analyzed in isolation.
Collapse
Affiliation(s)
- Stephen Treaster
- Department of Orthopaedics, Boston Children's Hospital, Boston, MA, United States.,Department of Genetics, Harvard Medical School, Boston, MA, United States
| | - David Karasik
- Azrieli Faculty of Medicine, Bar-Ilan University, Ramat Gan, Israel.,Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA, United States
| | - Matthew P Harris
- Department of Orthopaedics, Boston Children's Hospital, Boston, MA, United States.,Department of Genetics, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
14
|
Garagnani P, Marquis J, Delledonne M, Pirazzini C, Marasco E, Kwiatkowska KM, Iannuzzi V, Bacalini MG, Valsesia A, Carayol J, Raymond F, Ferrarini A, Xumerle L, Collino S, Mari D, Arosio B, Casati M, Ferri E, Monti D, Nacmias B, Sorbi S, Luiselli D, Pettener D, Castellani G, Sala C, Passarino G, De Rango F, D'Aquila P, Bertamini L, Martinelli N, Girelli D, Olivieri O, Giuliani C, Descombes P, Franceschi C. Whole-genome sequencing analysis of semi-supercentenarians. eLife 2021; 10:57849. [PMID: 33941312 PMCID: PMC8096429 DOI: 10.7554/elife.57849] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 04/09/2021] [Indexed: 12/13/2022] Open
Abstract
Extreme longevity is the paradigm of healthy aging as individuals who reached the extreme decades of human life avoided or largely postponed all major age-related diseases. In this study, we sequenced at high coverage (90X) the whole genome of 81 semi-supercentenarians and supercentenarians [105+/110+] (mean age: 106.6 ± 1.6) and of 36 healthy unrelated geographically matched controls (mean age 68.0 ± 5.9) recruited in Italy. The results showed that 105+/110+ are characterized by a peculiar genetic background associated with efficient DNA repair mechanisms, as evidenced by both germline data (common and rare variants) and somatic mutations patterns (lower mutation load if compared to younger healthy controls). Results were replicated in a second independent cohort of 333 Italian centenarians and 358 geographically matched controls. The genetics of 105+/110+ identified DNA repair and clonal haematopoiesis as crucial players for healthy aging and for the protection from cardiovascular events.
Collapse
Affiliation(s)
- 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, Stockholm, Sweden.,Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate), University of Bologna, Bologna, Italy
| | - Julien Marquis
- Nestlé Research, Société des Produits Nestlé SA, Lausanne, Switzerland
| | - Massimo Delledonne
- Functional Genomics Laboratory, Department of Biotechnology, University of Verona, Verona, Italy
| | - Chiara Pirazzini
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Elena Marasco
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy.,Applied Biomedical Research Center (CRBA), S. Orsola-Malpighi Polyclinic, Bologna, Italy
| | | | - Vincenzo Iannuzzi
- Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate), University of Bologna, Bologna, Italy
| | | | - Armand Valsesia
- Nestlé Research, Société des Produits Nestlé SA, Lausanne, Switzerland
| | - Jerome Carayol
- Nestlé Research, Société des Produits Nestlé SA, Lausanne, Switzerland
| | - Frederic Raymond
- Nestlé Research, Société des Produits Nestlé SA, Lausanne, Switzerland
| | - Alberto Ferrarini
- Functional Genomics Laboratory, Department of Biotechnology, University of Verona, Verona, Italy
| | - Luciano Xumerle
- Functional Genomics Laboratory, Department of Biotechnology, University of Verona, Verona, Italy
| | | | - Daniela Mari
- Fondazione Ca' Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Beatrice Arosio
- Fondazione Ca' Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy.,Geriatric Unit, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Martina Casati
- Fondazione Ca' Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Evelyn Ferri
- 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.,IRCCS Fondazione Don Carlo Gnocchi, Firenze, Italy
| | - Sandro Sorbi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy.,IRCCS Fondazione Don Carlo Gnocchi, Firenze, Italy
| | - Donata Luiselli
- Department for the Cultural Heritage (DBC), University of Bologna, Ravenna, 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
| | - Gastone Castellani
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Claudia Sala
- Department of Physics and Astronomy, University of Bologna, Bologna, Italy
| | - Giuseppe Passarino
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Francesco De Rango
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Patrizia D'Aquila
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Luca Bertamini
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy.,Department of Medicine, Unit of Internal Medicine, University of Verona, Verona, Italy
| | - Nicola Martinelli
- Department of Medicine, Unit of Internal Medicine, University of Verona, Verona, Italy
| | - Domenico Girelli
- Department of Medicine, Unit of Internal Medicine, University of Verona, Verona, Italy
| | - Oliviero Olivieri
- Department of Medicine, Unit of Internal Medicine, University of Verona, Verona, Italy
| | - 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, United Kingdom
| | - Patrick Descombes
- Nestlé Research, Société des Produits Nestlé SA, Lausanne, Switzerland
| | - Claudio Franceschi
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy.,IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy.,Department of Applied Mathematics and Laboratory of Systems Biology of Aging, Lobachevsky University, Nizhny Novgorod, Russian Federation
| |
Collapse
|
15
|
Dato S, Crocco P, De Rango F, Iannone F, Maletta R, Bruni AC, Saiardi A, Rose G, Passarino G. IP6K3 and IPMK variations in LOAD and longevity: Evidence for a multifaceted signaling network at the crossroad between neurodegeneration and survival. Mech Ageing Dev 2021; 195:111439. [PMID: 33497757 DOI: 10.1016/j.mad.2021.111439] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/23/2020] [Accepted: 01/18/2021] [Indexed: 12/18/2022]
Abstract
Several studies reported that genetic variants predisposing to neurodegeneration were at higher frequencies in centenarians than in younger controls, suggesting they might favor also longevity. IP6K3 and IPMK regulate many crucial biological functions by mediating synthesis of inositol poly- and pyrophosphates and by acting non-enzymatically via protein-protein interactions. Our previous studies suggested they affect Late Onset Alzheimer Disease (LOAD) and longevity, respectively. Here, in the same sample groups, we investigated whether variants of IP6K3 also affect longevity, and variants of IPMK also influence LOAD susceptibility. We found that: i) a SNP of IP6K3 previously associated with increased risk of LOAD increased the chance to become long-lived, ii) SNPs of IPMK, previously associated with decreased longevity, were protective factors for LOAD, as previously observed for UCP4. SNP-SNP interaction analysis, including our previous data, highlighted phenotype-specific interactions between sets of alleles. Moreover, linkage disequilibrium and eQTL data associated to analyzed variants suggested mitochondria as crossroad of interconnected pathways crucial for susceptibility to neurodegeneration and/or longevity. Overall, data support the view that in these traits interactions may be more important than single polymorphisms. This phenomenon may contribute to the non-additive heritability of neurodegeneration and longevity and be part of the missing heritability of these traits.
Collapse
Affiliation(s)
- Serena Dato
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy.
| | - Paolina Crocco
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy.
| | - Francesco De Rango
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy.
| | - Francesca Iannone
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy.
| | - Raffaele Maletta
- Regional Neurogenetic Centre, ASP Catanzaro, Lamezia Terme, Italy.
| | - Amalia C Bruni
- Regional Neurogenetic Centre, ASP Catanzaro, Lamezia Terme, Italy.
| | - Adolfo Saiardi
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, London, UK.
| | - Giuseppina Rose
- 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.
| |
Collapse
|
16
|
Nazarian A, Kulminski AM. Evaluation of the Genetic Variance of Alzheimer's Disease Explained by the Disease-Associated Chromosomal Regions. J Alzheimers Dis 2020; 70:907-915. [PMID: 31282417 DOI: 10.3233/jad-190168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Heritability analysis of complex traits/diseases is commonly performed to obtain illustrative information about the potential contribution of the genetic factors to their phenotypic variances. In this study, we investigated the narrow-sense heritability (h2) of Alzheimer's disease (AD) using genome-wide single-nucleotide polymorphisms (SNPs) data from three independent studies in the linear mixed models framework. Our meta-analyses demonstrated that the estimated h2 values (and their standard errors) of AD in liability scale were 0.280 (0.091), 0.348 (0.113), and 0.389 (0.126) assuming AD prevalence rates of 10%, 20%, or 30% at ages of 65+, 75+, and 85+ years, respectively. We also found that chromosomal regions containing two or more AD-associated SNPs at p < 5E-08 could collectively explain 37% of the additive genetic variance of AD in our samples. AD-associated regions in which at least one SNP had attained p < 5E-08 explained 56% of the additive genetic variance of AD. These regions harbored 3% and 11% of SNPs in our analyses. Also, the chromosomal regions containing two or more and one or more AD-associated SNPs at p < 5E-06 accounted for 72% and 94% of the additive genetic variance of AD, respectively. These regions harbored 27% and 44% of SNPs in our analyses. Our findings showed that the overall contribution of the additive genetic effects to the AD liability was moderate and age-dependent. Also, they supported the importance of focusing on known AD-associated chromosomal regions to investigate the genetic basis of AD, e.g., through haplotype analysis, analysis of heterogeneity, and functional studies.
Collapse
Affiliation(s)
- Alireza Nazarian
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, USA
| | - Alexander M Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, USA
| |
Collapse
|
17
|
Gutman D, Lidzbarsky G, Milman S, Gao T, Sin-Chan P, Gonzaga‐Jauregui C, Deelen J, Shuldiner AR, Barzilai N, Atzmon G. Similar burden of pathogenic coding variants in exceptionally long-lived individuals and individuals without exceptional longevity. Aging Cell 2020; 19:e13216. [PMID: 32860726 PMCID: PMC7576295 DOI: 10.1111/acel.13216] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 06/22/2020] [Accepted: 07/12/2020] [Indexed: 12/13/2022] Open
Abstract
Centenarians (exceptionally long‐lived individuals—ELLI) are a unique segment of the population, exhibiting long human lifespan and healthspan, despite generally practicing similar lifestyle habits as their peers. We tested disease‐associated mutation burden in ELLI genomes by determining the burden of pathogenic variants reported in the ClinVar and HGMD databases using data from whole exome sequencing (WES) conducted in a cohort of ELLI, their offspring, and control individuals without antecedents of familial longevity (n = 1879), all descendent from the founder population of Ashkenazi Jews. The burden of pathogenic variants did not differ between the three groups. Additional analyses of variants subtypes and variant effect predictor (VEP) biotype frequencies did not reveal a decrease of pathogenic or loss‐of‐function (LoF) variants in ELLI and offspring compared to the control group. Case–control pathogenic variants enrichment analyses conducted in ELLI and controls also did not identify significant differences in any of the variants between the groups and polygenic risk scores failed to provide a predictive model. Interestingly, cancer and Alzheimer's disease‐associated variants were significantly depleted in ELLI compared to controls, suggesting slower accumulation of mutation. That said, polygenic risk score analysis failed to find any predictive variants among the functional variants tested. The high similarity in the burden of pathogenic variation between ELLI and individuals without familial longevity supports the notion that extension of lifespan and healthspan in ELLI is not a consequence of pathogenic variant depletion but rather a result of other genomic, epigenomic, or potentially nongenomic properties.
Collapse
Affiliation(s)
- Danielle Gutman
- Faculty of Natural Sciences University of Haifa Haifa Israel
| | | | - Sofiya Milman
- Department of Medicine Albert Einstein College of Medicine Bronx New York USA
| | - Tina Gao
- Department of Medicine Albert Einstein College of Medicine Bronx New York USA
| | | | | | - Joris Deelen
- Max Planck Institute for Biology of Ageing Cologne Germany
- Molecular Epidemiology Department of Biochemical Data Sciences Leiden University Medical Center Leiden The Netherlands
| | | | - Nir Barzilai
- Department of Medicine Albert Einstein College of Medicine Bronx New York USA
- Genetic, Institute for Aging Research and the Diabetes Research Center Albert Einstein College of Medicine Bronx New York USA
| | - Gil Atzmon
- Faculty of Natural Sciences University of Haifa Haifa Israel
- Department of Medicine Albert Einstein College of Medicine Bronx New York USA
- Genetic, Institute for Aging Research and the Diabetes Research Center Albert Einstein College of Medicine Bronx New York USA
| | | |
Collapse
|
18
|
Abstract
The majority of research to understand the pathogenesis of and contributors to Alzheimer’s disease (AD) pathology, dementia, and disease progression has focused on studying individuals who have the disease or are at increased risk of having the disease. Yet there may be much to learn from individuals who have a paradoxical decreased risk of AD suggesting underlying protective factors. Centenarians demonstrate exceptional longevity that for a subset of the cohort is associated with an increased health span characterized by the delay or escape of age-related diseases including dementia. Here, I give evidence of the association of exceptional longevity with resistance and resilience to AD and describe how cohorts of centenarians and their offspring may serve as models of neuroprotection from AD. Discoveries of novel genetic, environmental, and behavioral factors that are associated with a decreased risk of AD may inform the development of interventions to slow or prevent AD in the general population. Centenarian cohorts may also be instrumental in serving as controls to individuals with AD to identify additional risk factors.
Collapse
|
19
|
Bosch-Boonstra-Schaaf optic atrophy syndrome (BBSOAS) initially diagnosed as ALG6-CDG: Functional evidence for benignity of the ALG6 c.391T>C (p.Tyr131His) variant and further expanding the BBSOAS phenotype. Eur J Med Genet 2020; 63:103941. [PMID: 32407885 DOI: 10.1016/j.ejmg.2020.103941] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/29/2020] [Accepted: 05/02/2020] [Indexed: 11/23/2022]
Abstract
Bosch-Boonstra-Schaaf optic atrophy syndrome (BBSOAS) is a recently described autosomal dominant syndrome of developmental delay, cortical vision loss with optic nerve atrophy, epilepsy, and autism spectrum disorder. Due to its many overlapping features with congenital disorders of glycosylation (CDG), the differential diagnosis between these disorders may be difficult and relies on molecular genetic testing. We report on a 31-year-old female initially diagnosed with ALG6-CDG based on glycosylation abnormalities on transferrin isoelectrofocusing and targeted genetic testing, and later diagnosed with BBSOAS by whole-exome sequencing (WES). Functional studies on cultured fibroblasts including Western blotting and RT-qPCR, as well as mass spectrometry of glycosylated transferrin and MALDI-TOF glycan analysis in serum, demonstrated normal glycosylation in this patient. In this report, we extend the phenotype of BBSOAS with ataxia and protein-losing enteropathy. This case is illustrative of the utility of whole exome sequencing in the diagnostic odyssey, and the potential pitfalls of relying on focused genetic testing results for diagnosis of conditions with complex overlapping phenotypes.
Collapse
|
20
|
Carlson P, Wojczynski MK, Druley T, Lee JH, Zmuda JM, Thyagarajan B. Prevalence of clinically actionable disease variants in exceptionally long-lived families. BMC Med Genomics 2020; 13:61. [PMID: 32272925 PMCID: PMC7146901 DOI: 10.1186/s12920-020-0710-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 03/27/2020] [Indexed: 12/15/2022] Open
Abstract
Background Phenotypic expression of pathogenic variants in individuals with no family history of inherited disorders remains unclear. Methods We evaluated the prevalence of pathogenic variants in 25 genes associated with Mendelian-inherited disorders in 3015 participants from 485 families in the Long Life Family Study (LLFS). Boot-strapping and Fisher’s exact test were used to determine whether allele frequencies in LLFS were significantly different from the allele frequencies reported in publicly available genomic databases. Results The proportions of pathogenic autosomal dominant mutation carriers in BRCA1 and SDHC in LLFS study participants were similar to those reported in publicly available genomic databases (0.03% vs. 0.0008%, p = 1 for BRCA1, and 0.08% vs. 0.003%, p = 0.05 for SDHC). The frequency of carriers of pathogenic autosomal recessive variants in CPT2, ACADM, SUMF1, WRN, ATM, and ACADVL were also similar in LLFS as compared to those reported in genomic databases. The lack of clinical disease among LLFS participants with well-established pathogenic variants in BRCA1 and SDHC suggests that penetrance of pathogenic variants may be different in long lived families. Conclusion Further research is needed to better understand the penetrance of pathogenic variants before expanding large scale genomic testing to asymptomatic individuals.
Collapse
Affiliation(s)
- Paige Carlson
- University of Minnesota Medical School, Duluth, MN, USA
| | - Mary K Wojczynski
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Todd Druley
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8116, St. Louis, MO, 63108, USA.,Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8116, St. Louis, MO, 63108, USA
| | - Joseph H Lee
- Sergievsky Center, College of Physicians and Surgeons, Columbia University New York, New York, NY, USA.,Taub Institute, College of Physicians and Surgeons, Columbia University New York, New York, NY, USA.,Departments of Epidemiology and Neurology, Columbia University New York, New York, NY, USA
| | - Joseph M Zmuda
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota, MMC 609, 420 Delaware street, Minneapolis, MN, 55455, USA.
| |
Collapse
|
21
|
Kulminski AM, Shu L, Loika Y, Nazarian A, Arbeev K, Ukraintseva S, Yashin A, Culminskaya I. APOE region molecular signatures of Alzheimer's disease across races/ethnicities. Neurobiol Aging 2020; 87:141.e1-141.e8. [PMID: 31813627 PMCID: PMC7064423 DOI: 10.1016/j.neurobiolaging.2019.11.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 08/19/2019] [Accepted: 11/06/2019] [Indexed: 11/20/2022]
Abstract
The role of even the strongest genetic risk factor for Alzheimer's disease (AD), the apolipoprotein E (APOE) ε4 allele, in its etiology remains poorly understood. We examined molecular signatures of AD defined as differences in linkage disequilibrium patterns between AD-affected and -unaffected whites (2673/16,246), Hispanics (392/867), and African Americans (285/1789), separately. We focused on 29 polymorphisms from 5 genes in the APOE region emphasizing beneficial and adverse effects of the APOE ε2- and ε4-coding single-nucleotide polymorphisms, respectively, and the differences in the linkage disequilibrium structures involving these alleles between AD-affected and -unaffected subjects. Susceptibility to AD is likely the result of complex interactions of the ε2 and ε4 alleles with other polymorphisms in the APOE region, and these interactions differ across races/ethnicities corroborating differences in the adverse and beneficial effects of the ε4 and ε2 alleles. Our findings support complex race/ethnicity-specific haplotypes promoting and protecting against AD in this region. They contribute to better understanding of polygenic and resilient mechanisms, which can explain why even homozygous ε4 carriers may not develop AD.
Collapse
Affiliation(s)
- Alexander M Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, USA.
| | - Leonardo Shu
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, USA
| | - Yury Loika
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, USA
| | - Alireza Nazarian
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, USA
| | - Konstantin Arbeev
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, USA
| | - Svetlana Ukraintseva
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, USA
| | - Anatoliy Yashin
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, USA
| | - Irina Culminskaya
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, USA
| |
Collapse
|
22
|
Earl J, Galindo-Pumariño C, Encinas J, Barreto E, Castillo ME, Pachón V, Ferreiro R, Rodríguez-Garrote M, González-Martínez S, Ramon Y Cajal T, Diaz LR, Chirivella-Gonzalez I, Rodriguez M, de Castro EM, García-Seisdedos D, Muñoz G, Rosa JMR, Marquez M, Malats N, Carrato A. A comprehensive analysis of candidate genes in familial pancreatic cancer families reveals a high frequency of potentially pathogenic germline variants. EBioMedicine 2020; 53:102675. [PMID: 32113160 PMCID: PMC7100610 DOI: 10.1016/j.ebiom.2020.102675] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 01/29/2020] [Accepted: 01/29/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The 5-year survival rate of patients with pancreatic ductal adenocarcinoma (PDAC) is around 5% due to the fact that the majority of patients present with advanced disease that is treatment resistant. Familial pancreatic cancer (FPC) is a rare disorder that is defined as a family with at least two affected first degree relatives, with an estimated incidence of 4%-10%. The genetic basis is unknown in the majority of families although around 10%-13% of families carry germline mutations in known genes associated with hereditary cancer and pancreatitis syndromes. METHODS Panel sequencing was performed of 35 genes associated with hereditary cancer in 43 PDAC cases from families with an apparent hereditary pancreatic cancer syndrome. FINDINGS Pathogenic variants were identified in 19% (5/26) of PDAC cases from pure FPC families in the genes MLH1, CDKN2A, POLQ and FANCM. Low frequency potentially pathogenic VUS were also identified in 35% (9/26) of PDAC cases from FPC families in the genes FANCC, MLH1, PMS2, CFTR, APC and MUTYH. Furthermore, an important proportion of PDAC cases harboured more than one pathogenic, likely pathogenic or potentially pathogenic VUS, highlighting the multigene phenotype of FPC. INTERPRETATION The genetic basis of familial or hereditary pancreatic cancer can be explained in 21% of families by previously described hereditary cancer genes. Low frequency variants in other DNA repair genes are also present in 35% of families which may contribute to the risk of pancreatic cancer development. FUNDING This study was funded by the Instituto de Salud Carlos III (Plan Estatal de I + D + i 2013-2016): ISCIII (PI09/02221, PI12/01635, PI15/02101 and PI18/1034) and co-financed by the European Development Regional Fund ''A way to achieve Europe'' (ERDF), the Biomedical Research Network in Cancer: CIBERONC (CB16/12/00446), Red Temática de investigación cooperativa en cáncer: RTICC (RD12/0036/0073) and La Asociación Española contra el Cáncer: AECC (Grupos Coordinados Estables 2016).
Collapse
Affiliation(s)
- Julie Earl
- Molecular Epidemiology and Predictive Tumor Markers Group, Medical Oncology Research Laboratory, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9100, 28034 Madrid, Spain; Biomedical Research Network in Cancer (CIBERONC), C/Monforte de Lemos 3-5, Pabellón 11, 28029 Madrid, Spain.
| | - Cristina Galindo-Pumariño
- Molecular Epidemiology and Predictive Tumor Markers Group, Medical Oncology Research Laboratory, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9100, 28034 Madrid, Spain; Biomedical Research Network in Cancer (CIBERONC), C/Monforte de Lemos 3-5, Pabellón 11, 28029 Madrid, Spain
| | - Jessica Encinas
- Molecular Epidemiology and Predictive Tumor Markers Group, Medical Oncology Research Laboratory, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9100, 28034 Madrid, Spain
| | - Emma Barreto
- Molecular Epidemiology and Predictive Tumor Markers Group, Medical Oncology Research Laboratory, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9100, 28034 Madrid, Spain
| | - Maria E Castillo
- Molecular Epidemiology and Predictive Tumor Markers Group, Medical Oncology Research Laboratory, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9100, 28034 Madrid, Spain
| | - Vanessa Pachón
- Molecular Epidemiology and Predictive Tumor Markers Group, Medical Oncology Research Laboratory, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9100, 28034 Madrid, Spain; Biomedical Research Network in Cancer (CIBERONC), C/Monforte de Lemos 3-5, Pabellón 11, 28029 Madrid, Spain
| | - Reyes Ferreiro
- Molecular Epidemiology and Predictive Tumor Markers Group, Medical Oncology Research Laboratory, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9100, 28034 Madrid, Spain
| | - Mercedes Rodríguez-Garrote
- Molecular Epidemiology and Predictive Tumor Markers Group, Medical Oncology Research Laboratory, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9100, 28034 Madrid, Spain; Biomedical Research Network in Cancer (CIBERONC), C/Monforte de Lemos 3-5, Pabellón 11, 28029 Madrid, Spain
| | - Silvia González-Martínez
- Molecular Epidemiology and Predictive Tumor Markers Group, Medical Oncology Research Laboratory, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9100, 28034 Madrid, Spain
| | - Teresa Ramon Y Cajal
- Medical Oncology Department, Santa Creu i Sant Pau Hospital, Mas Casanovas, 90, 08041 Barcelona, Spain.
| | - Luis Robles Diaz
- Familial and Hereditary Cancer Unit. Medical Oncology Department, 12 de Octubre Hospital, Av. Cordoba, s/n, 28041 Madrid, Spain.
| | - Isabel Chirivella-Gonzalez
- Genetic Counselling Unit, Valencia University Hospital Clinic, Av. de Blasco Ibáñez, 17, 46010 Valencia, Spain.
| | - Montse Rodriguez
- A Coruña Biomedical Research Institute, Hospital Teresa Herrera, Xubias de Arriba, 84, 15006 A Coruña, Spain.
| | - Eva Martínez de Castro
- Medical Oncology Department, Marqués de Valdecilla University Hospital, Av. Valdecilla, 25, 39008 Santander, Spain.
| | - David García-Seisdedos
- Translational Genomics Core Facility, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Gloria Muñoz
- Translational Genomics Core Facility, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Juan Manuel Rosa Rosa
- Pathology Department, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Mirari Marquez
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain; Biomedical Research Network in Cancer (CIBERONC), C/Monforte de Lemos 3-5, Pabellón 11, 28029 Madrid, Spain
| | - Nuría Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain; Biomedical Research Network in Cancer (CIBERONC), C/Monforte de Lemos 3-5, Pabellón 11, 28029 Madrid, Spain.
| | - Alfredo Carrato
- Molecular Epidemiology and Predictive Tumor Markers Group, Medical Oncology Research Laboratory, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9100, 28034 Madrid, Spain; Biomedical Research Network in Cancer (CIBERONC), C/Monforte de Lemos 3-5, Pabellón 11, 28029 Madrid, Spain.
| |
Collapse
|
23
|
Kulminski AM, Shu L, Loika Y, He L, Nazarian A, Arbeev K, Ukraintseva S, Yashin A, Culminskaya I. Genetic and regulatory architecture of Alzheimer's disease in the APOE region. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2020; 12:e12008. [PMID: 32211503 PMCID: PMC7085286 DOI: 10.1002/dad2.12008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 11/07/2019] [Accepted: 11/20/2019] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Apolipoprotein E (APOE) ε2 and ε4 alleles encoded by rs7412 and rs429358 polymorphisms, respectively, are landmark contra and pro "risk" factors for Alzheimer's disease (AD). METHODS We examined differences in linkage disequilibrium (LD) structures between (1) AD-affected and unaffected subjects and (2) older AD-unaffected and younger subjects in the 19q13.3 region harboring rs7412 and rs429358. RESULTS AD is associated with sex-nonspecific heterogeneous patterns of decreased and increased LD of rs7412 and rs429358, respectively, with other polymorphisms from five genes in this region in AD-affected subjects. The LD patterns in older AD-unaffected subjects resembled those in younger individuals. Polarization of the ε4- and ε2 allele-related heterogeneous LD clusters differentiated cell types and implicated specific tissues in AD pathogenesis. DISCUSSION Protection and predisposition to AD is characterized by an interplay of rs7412 and rs429358, with multiple polymorphisms in the 19q13.3 region in a tissue-specific manner, which is not driven by common evolutionary forces.
Collapse
Affiliation(s)
- Alexander M. Kulminski
- Biodemography of Aging Research UnitSocial Science Research InstituteDuke UniversityDurhamNorth Carolina
| | - Leonardo Shu
- Biodemography of Aging Research UnitSocial Science Research InstituteDuke UniversityDurhamNorth Carolina
| | - Yury Loika
- Biodemography of Aging Research UnitSocial Science Research InstituteDuke UniversityDurhamNorth Carolina
| | - Liang He
- Biodemography of Aging Research UnitSocial Science Research InstituteDuke UniversityDurhamNorth Carolina
| | - Alireza Nazarian
- Biodemography of Aging Research UnitSocial Science Research InstituteDuke UniversityDurhamNorth Carolina
| | - Konstantin Arbeev
- Biodemography of Aging Research UnitSocial Science Research InstituteDuke UniversityDurhamNorth Carolina
| | - Svetlana Ukraintseva
- Biodemography of Aging Research UnitSocial Science Research InstituteDuke UniversityDurhamNorth Carolina
| | - Anatoliy Yashin
- Biodemography of Aging Research UnitSocial Science Research InstituteDuke UniversityDurhamNorth Carolina
| | - Irina Culminskaya
- Biodemography of Aging Research UnitSocial Science Research InstituteDuke UniversityDurhamNorth Carolina
| |
Collapse
|
24
|
Gutman D, Rivkin E, Fadida A, Sharvit L, Hermush V, Rubin E, Kirshner D, Sabin I, Dwolatzky T, Atzmon G. Exceptionally Long-Lived Individuals (ELLI) Demonstrate Slower Aging Rate Calculated by DNA Methylation Clocks as Possible Modulators for Healthy Longevity. Int J Mol Sci 2020; 21:ijms21020615. [PMID: 31963520 PMCID: PMC7013521 DOI: 10.3390/ijms21020615] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 01/14/2020] [Accepted: 01/15/2020] [Indexed: 12/23/2022] Open
Abstract
Exceptionally long-lived individuals (ELLI) who are the focus of many healthy longevity studies around the globe are now being studied in Israel. The Israeli Multi-Ethnic Centenarian Study (IMECS) cohort is utilized here for assessment of various DNA methylation clocks. Thorough phenotypic characterization and whole blood samples were obtained from ELLI, offspring of ELLI, and controls aged 53–87 with no familial exceptional longevity. DNA methylation was assessed using Illumina MethylationEPIC Beadchip and applied to DNAm age online tool for age and telomere length predictions. Relative telomere length was assessed using qPCR T/S (Telomere/Single copy gene) ratios. ELLI demonstrated juvenile performance in DNAm age clocks and overall methylation measurement, with preserved cognition and relative telomere length. Our findings suggest a favorable DNA methylation profile in ELLI enabling a slower rate of aging in those individuals in comparison to controls. It is possible that DNA methylation is a key modulator of the rate of aging and thus the ELLI DNAm profile promotes healthy longevity.
Collapse
Affiliation(s)
- Danielle Gutman
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel; (D.G.); (L.S.)
| | - Elina Rivkin
- Faculty of Public Health, University of Haifa, Haifa 3498838, Israel; (E.R.); (A.F.)
| | - Almog Fadida
- Faculty of Public Health, University of Haifa, Haifa 3498838, Israel; (E.R.); (A.F.)
| | - Lital Sharvit
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel; (D.G.); (L.S.)
| | - Vered Hermush
- Department of Geriatrics and Skilled Nursing, Laniado Medical Center, Netanya 4244916, Israel;
- Ruth and Bruce Rappaport Faculty of Medicine, Technion–Israel Institute of Technology, Haifa 3200003, Israel; (D.K.); (I.S.); (T.D.)
| | - Elad Rubin
- Department of Geriatrics, Rambam Health Care Campus, Haifa 3109601, Israel;
| | - Dani Kirshner
- Ruth and Bruce Rappaport Faculty of Medicine, Technion–Israel Institute of Technology, Haifa 3200003, Israel; (D.K.); (I.S.); (T.D.)
- Department of Geriatrics, Rambam Health Care Campus, Haifa 3109601, Israel;
| | - Irina Sabin
- Ruth and Bruce Rappaport Faculty of Medicine, Technion–Israel Institute of Technology, Haifa 3200003, Israel; (D.K.); (I.S.); (T.D.)
- Department of Geriatrics, Rambam Health Care Campus, Haifa 3109601, Israel;
| | - Tzvi Dwolatzky
- Ruth and Bruce Rappaport Faculty of Medicine, Technion–Israel Institute of Technology, Haifa 3200003, Israel; (D.K.); (I.S.); (T.D.)
- Department of Geriatrics, Rambam Health Care Campus, Haifa 3109601, Israel;
| | - Gil Atzmon
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel; (D.G.); (L.S.)
- Departments of Genetics and Medicine, Division of endocrinology, Institute for Aging Research and the Diabetes Research Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
- Correspondence: ; Tel.: +972-4664-7927
| |
Collapse
|
25
|
Finch CE, Kulminski AM. The Alzheimer's Disease Exposome. Alzheimers Dement 2019; 15:1123-1132. [PMID: 31519494 PMCID: PMC6788638 DOI: 10.1016/j.jalz.2019.06.3914] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 06/06/2019] [Accepted: 06/12/2019] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Environmental factors are poorly understood in the etiology of Alzheimer's disease (AD) and related dementias. The importance of environmental factors in gene environment interactions (GxE) is suggested by wide individual differences in cognitive loss, even for carriers of AD-risk genetic variants. RESULTS AND DISCUSSION We propose the "AD exposome" to comprehensively assess the modifiable environmental factors relevant to genetic underpinnings of cognitive aging and AD. Analysis of endogenous and exogenous environmental factors requires multi-generational consideration of these interactions over age and time (GxExT). New computational approaches to the multi-level complexities may identify accessible interventions for individual brain aging. International collaborations on diverse populations are needed to identify the most relevant exposures over the life course for GxE interactions.
Collapse
Affiliation(s)
- Caleb E Finch
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA.
| | - Alexander M Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, USA.
| |
Collapse
|
26
|
Marinko J, Huang H, Penn WD, Capra JA, Schlebach JP, Sanders CR. Folding and Misfolding of Human Membrane Proteins in Health and Disease: From Single Molecules to Cellular Proteostasis. Chem Rev 2019; 119:5537-5606. [PMID: 30608666 PMCID: PMC6506414 DOI: 10.1021/acs.chemrev.8b00532] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Indexed: 12/13/2022]
Abstract
Advances over the past 25 years have revealed much about how the structural properties of membranes and associated proteins are linked to the thermodynamics and kinetics of membrane protein (MP) folding. At the same time biochemical progress has outlined how cellular proteostasis networks mediate MP folding and manage misfolding in the cell. When combined with results from genomic sequencing, these studies have established paradigms for how MP folding and misfolding are linked to the molecular etiologies of a variety of diseases. This emerging framework has paved the way for the development of a new class of small molecule "pharmacological chaperones" that bind to and stabilize misfolded MP variants, some of which are now in clinical use. In this review, we comprehensively outline current perspectives on the folding and misfolding of integral MPs as well as the mechanisms of cellular MP quality control. Based on these perspectives, we highlight new opportunities for innovations that bridge our molecular understanding of the energetics of MP folding with the nuanced complexity of biological systems. Given the many linkages between MP misfolding and human disease, we also examine some of the exciting opportunities to leverage these advances to address emerging challenges in the development of therapeutics and precision medicine.
Collapse
Affiliation(s)
- Justin
T. Marinko
- Department
of Biochemistry, Vanderbilt University, Nashville, Tennessee 37240, United States
- Center
for Structural Biology, Vanderbilt University, Nashville, Tennessee 37240, United States
| | - Hui Huang
- Department
of Biochemistry, Vanderbilt University, Nashville, Tennessee 37240, United States
- Center
for Structural Biology, Vanderbilt University, Nashville, Tennessee 37240, United States
| | - Wesley D. Penn
- Department
of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - John A. Capra
- Center
for Structural Biology, Vanderbilt University, Nashville, Tennessee 37240, United States
- Department
of Biological Sciences, Vanderbilt University, Nashville, Tennessee 37245, United States
| | - Jonathan P. Schlebach
- Department
of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Charles R. Sanders
- Department
of Biochemistry, Vanderbilt University, Nashville, Tennessee 37240, United States
| |
Collapse
|
27
|
Puca AA, Spinelli C, Accardi G, Villa F, Caruso C. Centenarians as a model to discover genetic and epigenetic signatures of healthy ageing. Mech Ageing Dev 2018; 174:95-102. [DOI: 10.1016/j.mad.2017.10.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 10/26/2017] [Accepted: 10/28/2017] [Indexed: 01/07/2023]
|
28
|
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: 1.2] [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
|
29
|
Freudenberg-Hua Y, Li W, Davies P. The Role of Genetics in Advancing Precision Medicine for Alzheimer's Disease-A Narrative Review. Front Med (Lausanne) 2018; 5:108. [PMID: 29740579 PMCID: PMC5928202 DOI: 10.3389/fmed.2018.00108] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 04/03/2018] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is the most common type of dementia, which has a substantial genetic component. AD affects predominantly older people. Accordingly, the prevalence of dementia has been rising as the population ages. To date, there are no effective interventions that can cure or halt the progression of AD. The only available treatments are the management of certain symptoms and consequences of dementia. The current state-of-the-art medical care for AD comprises three simple principles: prevent the preventable, achieve early diagnosis, and manage the manageable symptoms. This review provides a summary of the current state of knowledge of risk factors for AD, biological diagnostic testing, and prospects for treatment. Special emphasis is given to recent advances in genetics of AD and the way genomic data may support prevention, early intervention, and development of effective pharmacological treatments. Mutations in the APP, PSEN1, and PSEN2 genes cause early onset Alzheimer's disease (EOAD) that follows a Mendelian inheritance pattern. For late onset Alzheimer's disease (LOAD), APOE4 was identified as a major risk allele more than two decades ago. Population-based genome-wide association studies of late onset AD have now additionally identified common variants at roughly 30 genetic loci. Furthermore, rare variants (allele frequency <1%) that influence the risk for LOAD have been identified in several genes. These genetic advances have broadened our insights into the biological underpinnings of AD. Moreover, the known genetic risk variants could be used to identify presymptomatic individuals at risk for AD and support diagnostic assessment of symptomatic subjects. Genetic knowledge may also facilitate precision medicine. The goal of precision medicine is to use biological knowledge and other health information to predict individual disease risk, understand disease etiology, identify disease subcategories, improve diagnosis, and provide personalized treatment strategies. We discuss the potential role of genetics in advancing precision medicine for AD along with its ethical challenges. We outline strategies to implement genomics into translational clinical research that will not only improve accuracy of dementia diagnosis, thus enabling more personalized treatment strategies, but may also speed up the discovery of novel drugs and interventions.
Collapse
Affiliation(s)
- Yun Freudenberg-Hua
- Litwin-Zucker Center for the study of Alzheimer’s Disease, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
- Division of Geriatric Psychiatry, Zucker Hillside Hospital, Northwell Health, Glen Oaks, NY, United States
| | - Wentian Li
- Robert S Boas Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Peter Davies
- Litwin-Zucker Center for the study of Alzheimer’s Disease, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
| |
Collapse
|
30
|
Louwersheimer E, Cohn-Hokke PE, Pijnenburg YAL, Weiss MM, Sistermans EA, Rozemuller AJ, Hulsman M, van Swieten JC, van Duijn CM, Barkhof F, Koene T, Scheltens P, Van der Flier WM, Holstege H. Rare Genetic Variant in SORL1 May Increase Penetrance of Alzheimer's Disease in a Family with Several Generations of APOE-ɛ4 Homozygosity. J Alzheimers Dis 2018; 56:63-74. [PMID: 27911290 PMCID: PMC5240543 DOI: 10.3233/jad-160091] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Background: The major genetic risk factor for late onset Alzheimer’s disease (AD) is the APOE-ɛ4 allele. However, APOE-ɛ4 homozygosity is not fully penetrant, suggesting co-occurrence of additional genetic variants. Objective: To identify genetic factors that, next to APOE-ɛ4 homozygosity, contribute to the development of AD. Methods: We identified a family with nine AD patients spanning four generations, with an inheritance pattern suggestive of autosomal dominant AD, with no variants in PSEN1, PSEN2, or APP. We collected DNA from four affected and seven unaffected family members and performed exome sequencing on DNA from three affected and one unaffected family members. Results: All affected family members were homozygous for the APOE-ɛ4 allele. Statistical analysis revealed that AD onset in this family was significantly earlier than could be expected based on APOE genotype and gender. Next to APOE-ɛ4 homozygosity, we found that all four affected family members carried a rare variant in the VPS10 domain of the SORL1 gene, associated with AβPP processing and AD risk. Furthermore, three of four affected family members carried a rare variant in the TSHZ3 gene, also associated with AβPP processing. Affected family members presented between 61 and 74 years, with variable presence of microbleeds/cerebral amyloid angiopathy and electroencephalographic abnormalities. Conclusion: We hypothesize that next to APOE-ɛ4 homozygosity, impaired SORL1 protein function, and possibly impaired TSHZ3 function, further disturbed Aβ processing. The convergence of these genetic factors over several generations might clarify the increased AD penetrance and the autosomal dominant-like inheritance pattern of AD as observed in this family.
Collapse
Affiliation(s)
- Eva Louwersheimer
- Alzheimer Center and Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Petra E Cohn-Hokke
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands
| | - Yolande A L Pijnenburg
- Alzheimer Center and Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Marjan M Weiss
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands
| | - Erik A Sistermans
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Marc Hulsman
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands.,Delft Bioinformatics Laboratory, Delft University of Technology, Delft, The Netherlands
| | - John C van Swieten
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands.,Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Cock M van Duijn
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands.,Institutes of Neurology and Healthcare Engineering, UCL, London, UK
| | - Teddy Koene
- Alzheimer Center & Department of Medical Psychology, VU University Medical Center, Amsterdam, The Netherlands
| | - Philip Scheltens
- Alzheimer Center and Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Wiesje M Van der Flier
- Alzheimer Center and Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands.,Department of Epidemiology & Biostatistics, VU University Medical Center, Amsterdam, The Netherlands
| | - Henne Holstege
- Alzheimer Center and Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands.,Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
31
|
Snezhkina AV, Lukyanova EN, Kalinin DV, Pokrovsky AV, Dmitriev AA, Koroban NV, Pudova EA, Fedorova MS, Volchenko NN, Stepanov OA, Zhevelyuk EA, Kharitonov SL, Lipatova AV, Abramov IS, Golovyuk AV, Yegorov YE, Vishnyakova KS, Moskalev AA, Krasnov GS, Melnikova NV, Shcherbo DS, Kiseleva MV, Kaprin AD, Alekseev BY, Zaretsky AR, Kudryavtseva AV. Exome analysis of carotid body tumor. BMC Med Genomics 2018; 11:17. [PMID: 29504908 PMCID: PMC5836820 DOI: 10.1186/s12920-018-0327-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Carotid body tumor (CBT) is a form of head and neck paragangliomas (HNPGLs) arising at the bifurcation of carotid arteries. Paragangliomas are commonly associated with germline and somatic mutations involving at least one of more than thirty causative genes. However, the specific functionality of a number of these genes involved in the formation of paragangliomas has not yet been fully investigated. Methods Exome library preparation was carried out using Nextera® Rapid Capture Exome Kit (Illumina, USA). Sequencing was performed on NextSeq 500 System (Illumina). Results Exome analysis of 52 CBTs revealed potential driver mutations (PDMs) in 21 genes: ARNT, BAP1, BRAF, BRCA1, BRCA2, CDKN2A, CSDE1, FGFR3, IDH1, KIF1B, KMT2D, MEN1, RET, SDHA, SDHB, SDHC, SDHD, SETD2, TP53BP1, TP53BP2, and TP53I13. In many samples, more than one PDM was identified. There are also 41% of samples in which we did not identify any PDM; in these cases, the formation of CBT was probably caused by the cumulative effect of several not highly pathogenic mutations. Estimation of average mutation load demonstrated 6–8 mutations per megabase (Mb). Genes with the highest mutation rate were identified. Conclusions Exome analysis of 52 CBTs for the first time revealed the average mutation load for these tumors and also identified potential driver mutations as well as their frequencies and co-occurrence with the other PDMs. Electronic supplementary material The online version of this article (10.1186/s12920-018-0327-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Elena N Lukyanova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Dmitry V Kalinin
- Vishnevsky Institute of Surgery, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Anatoly V Pokrovsky
- Vishnevsky Institute of Surgery, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Alexey A Dmitriev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Nadezhda V Koroban
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Elena A Pudova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Maria S Fedorova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Nadezhda N Volchenko
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Oleg A Stepanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Ekaterina A Zhevelyuk
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Sergey L Kharitonov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Anastasiya V Lipatova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Ivan S Abramov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Alexander V Golovyuk
- Vishnevsky Institute of Surgery, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Yegor E Yegorov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Khava S Vishnyakova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Alexey A Moskalev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - George S Krasnov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Nataliya V Melnikova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Dmitry S Shcherbo
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - Marina V Kiseleva
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Andrey D Kaprin
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Boris Y Alekseev
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Andrew R Zaretsky
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - Anna V Kudryavtseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia. .,National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia.
| |
Collapse
|
32
|
Maher BS, Latendresse S, Vanyukov MM. Informing Prevention and Intervention Policy Using Genetic Studies of Resistance. PREVENTION SCIENCE : THE OFFICIAL JOURNAL OF THE SOCIETY FOR PREVENTION RESEARCH 2018; 19:49-57. [PMID: 27943075 PMCID: PMC5466512 DOI: 10.1007/s11121-016-0730-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The common paradigm for conceptualizing the influence of genetic and environmental factors on a particular disease relies on the concept of risk. Consequently, the bulk of etiologic, including genetic, work focuses on "risk" factors. These factors are aggregated at the high end of the distribution of liability to disease, the latent variable underlying the distribution of probability and severity of a disorder. However, liability has a symmetric but distinct aspect to risk, resistance to disorder. Resistance factors, aggregated at the low end of the liability distribution and supporting health and recovery, appear to be more promising for effective prevention and intervention. Herein, we discuss existing work on resistance factors, highlighting those with known genetic influences. We examine the utility of incorporating resistance genetics in prevention and intervention trials and compare the statistical power of a series of ascertainment schemes to develop a general framework for examining resistance outcomes in genetically informative designs. We find that an approach that samples individuals discordant on measured liability, a low-risk design, is the most feasible design and yields power equivalent to or higher than commonly used designs for detecting resistance genetic and environmental effects.
Collapse
Affiliation(s)
- Brion S Maher
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, 624 N. Broadway Ave., Baltimore, MD, 21205, USA.
| | - Shawn Latendresse
- Department of Psychology and Neuroscience, Baylor University, Waco, TX, USA
| | - Michael M Vanyukov
- Departments of Pharmaceutical Sciences, Psychiatry, and Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
33
|
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: 72] [Impact Index Per Article: 10.3] [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
|
34
|
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: 6.6] [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]
|
35
|
De Luca V, Orfei MD, Gaudenzi S, Caltagirone C, Spalletta G. Inverse effect of the APOE epsilon4 allele in late- and early-onset Alzheimer's disease. Eur Arch Psychiatry Clin Neurosci 2016; 266:599-606. [PMID: 26714935 DOI: 10.1007/s00406-015-0663-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 11/23/2015] [Indexed: 12/22/2022]
Abstract
In Alzheimer's disease patients (AD), the age at onset (AAO) ranges from 40 to 90. Usually, AD patients who develop symptoms before the age of 65 are classified as early onset (EO). The best known genetic risk factor for AD is the ε4 allele of the apolipoprotein E (APOE). In this study, 474 subjects with AD were consecutively recruited in the memory clinic of the Santa Lucia Foundation in Rome. The best fitting model for the discrimination between EO and late onset (LO) was chosen based on lowest value of the Bayesian Information Criterion, which suggests the theoretical model with minimal deviation from the empirical distribution function of AAO in our sample. The FMM was used to compare EO and LO groups with respect to the following demographic and clinical variables: gender, age, education, MMSE and NPI. Furthermore a quantitative assessment of ADL and IADL was performed. Finally, the frequency of the APOE ε4 allele was compared in EO and LO groups. Using the admixture analysis, we established that the AAO discriminating EO from LO-AD was 63-64. Higher education was associated with earlier onset in the EO but not in LO, and duration of illness was associated with earlier onset only in LO. The ε4 allele was associated with later onset in EO but earlier onset in LO. Finally, increased impairment in ADL, IADL and NPI was associated with later onset only in the LO subgroup. Thus, the ε4 allele of the APOE gene was significantly associated with both EO and LO distributions but with opposite effect, suggesting genetic heterogeneity. Additional studies are needed to further clarify the genetic mechanisms differentiating EO- and LO-AD.
Collapse
Affiliation(s)
- Vincenzo De Luca
- Centre for Addiction and Mental Health (CAMH), EEG and Genetics Lab, Department of Psychiatry, University of Toronto, 250 College Street, Room R340, Toronto, ON, M5T 1R8, USA.
| | - Maria Donata Orfei
- Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Sara Gaudenzi
- Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Carlo Caltagirone
- Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Gianfranco Spalletta
- Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Rome, Italy
| |
Collapse
|
36
|
Kato K, Zweig R, Schechter CB, Barzilai N, Atzmon G. Positive attitude toward life, emotional expression, self-rated health, and depressive symptoms among centenarians and near-centenarians. Aging Ment Health 2016; 20:930-9. [PMID: 26114814 PMCID: PMC5048681 DOI: 10.1080/13607863.2015.1056770] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
OBJECTIVES Favorable attitudes, emotions, personality characteristics, and self-rated health have been associated with successful aging in late life. However, less is known regarding these constructs and their relationships to mental health outcomes in the oldest old persons. This study examined cross-sectional relationships of these psychological factors to depressive symptoms in centenarians and near-centenarians. METHODS A selected sample of Ashkenazi Jewish older adults aged 98-107 (n = 54, 78% female) without significant cognitive impairment participated. Cognitive function was assessed by Mini-Mental Status Examination, positive attitude toward life and emotional expression by the Personality Outlook Profile Scale (POPS), self-rated health by participants' subjective rating of their present health, and depressive symptoms by the Geriatric Depression Scale. RESULTS Results demonstrated inverse associations of the positive attitude toward life domain of the POPS and self-rated health with participants' levels of depressive symptoms even after adjusting for the effects of history of medical illnesses, cognitive function, and demographic variables. Additionally, participants with high levels of care showed higher levels of depressive symptoms. Path analysis supported the partially mediating role of positive attitude toward life in the relationship between self-rated health and depressive symptoms. CONCLUSION These findings emphasized the important roles of positive attitudes and emotions as well as self-rated health in mental health outcomes in the oldest old. Although, limited by its cross-sectional design, findings suggest these psychological factors may exert protective effects on mental health outcomes in advanced age.
Collapse
Affiliation(s)
- Kaori Kato
- Woodhull Medical and Mental Health Center / NYU Langone Medical School, Department of Psychiatry, 760 Broadway, Brooklyn, NY 11206, USA
| | - Richard Zweig
- Ferkauf Graduate School of Psychology, Yeshiva University, 1300 Morris Park Avenue, Bronx, NY 10461, USA. (718)430-3958.
| | - Clyde B. Schechter
- Department of Family and Social Medicine and Epidemiology and Population Health, Albert Einstein College of Medicine, Yeshiva University, 1300 Morris Park Avenue, Bronx, NY 10461, USA. (718)430-2754.
| | - Nir Barzilai
- Departments of Medicine and Genetics, Albert Einstein College of Medicine, Yeshiva University, 1300 Morris Park Avenue, Bronx, NY 10461, USA. (718)430-3144.
| | - Gil Atzmon
- Departments of Medicine and Genetics, Albert Einstein College of Medicine, Yeshiva University, 1300 Morris Park Avenue, Bronx, NY 10461, USA. (718)430-3628.
| |
Collapse
|
37
|
Ismail K, Nussbaum L, Sebastiani P, Andersen S, Perls T, Barzilai N, Milman S. Compression of Morbidity Is Observed Across Cohorts with Exceptional Longevity. J Am Geriatr Soc 2016; 64:1583-91. [PMID: 27377170 PMCID: PMC4988893 DOI: 10.1111/jgs.14222] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
OBJECTIVES To determine, in a sample of Ashkenazi Jewish aged 95 and older, whether there is a compression of morbidity similar to what has been reported in other cohorts with exceptional longevity. DESIGN Case-control study. SETTING Longevity Genes Project (LGP) and New England Centenarian Study (NECS). PARTICIPANTS LGP (n = 439, mean age 97.8 ± 2.8) and NECS (n = 1,498, mean age 101.4 ± 4.0) participants with exceptional longevity and their respective younger referent cohorts (LGP, n = 696; NECS, n = 302). MEASUREMENTS Self- and proxy reports of age of onset of cancer, cardiovascular disease, diabetes mellitus, hypertension, osteoporosis, and stroke. RESULTS Long-lived individuals from LGP and NECS had later age of onset of cancer, cardiovascular disease, diabetes mellitus, hypertension, and osteoporosis than their respective younger reference groups. The risk of overall morbidity was lower in participants with exceptional longevity than in younger participants (NECS men: relative risk (RR) = 0.12, women: RR = 0.20; LGP men: RR = 0.18, women: RR = 0.24). The age at which 20% of each of the groups with exceptional longevity experienced specific diseases was between 18 and 24 years later than in the reference groups, stratified according to sex. CONCLUSION The similar extension of health span and compression of morbidity seen in NECS and LGP participants with exceptional longevity further validates the utility of these rare individuals for the study of factors that delay or prevent a broad spectrum of diseases otherwise associated with mortality and disability.
Collapse
Affiliation(s)
- Khadija Ismail
- Department of Medicine, Division of Endocrinology and Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Lisa Nussbaum
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118
| | - Paola Sebastiani
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118
| | - Stacy Andersen
- Geriatrics Section, Department of Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA 02118
| | - Thomas Perls
- Geriatrics Section, Department of Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA 02118
| | - Nir Barzilai
- Department of Medicine, Division of Endocrinology and Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Sofiya Milman
- Department of Medicine, Division of Endocrinology and Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461
| |
Collapse
|
38
|
Freudenberg-Hua Y, Li W, Abhyankar A, Vacic V, Cortes V, Ben-Avraham D, Koppel J, Greenwald B, Germer S, Darnell RB, Barzilai N, Freudenberg J, Atzmon G, Davies P. Differential burden of rare protein truncating variants in Alzheimer's disease patients compared to centenarians. Hum Mol Genet 2016; 25:3096-3105. [PMID: 27260402 DOI: 10.1093/hmg/ddw150] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 05/06/2016] [Accepted: 05/13/2016] [Indexed: 12/31/2022] Open
Abstract
We compared coding region variants of 53 cognitively healthy centenarians and 45 patients with Alzheimer's disease (AD), all of Ashkenazi Jewish (AJ) ancestry. Despite the small sample size, the known AD risk variant APOE4 reached genome-wide significance, indicating the advantage of utilizing 'super-controls'. We restricted our subsequent analysis to rare variants observed at most once in the 1000 Genomes database and having a minor allele frequency below 2% in our AJ sample. We compared the burden of predicted protein altering variants between cases and controls as normalized by the level of rare synonymous variants. We observed an increased burden among AD subjects for predicted loss-of-function (LoFs) variants defined as stop-gain, frame shift, initiation codon (INIT) and splice site mutations (n = 930, OR = 1.3, P = 1.5×E-5). There was no enrichment across all rare protein altering variants defined as missense plus LoFs, in frame indels and stop-loss variants (n = 13 014, OR = 0.97, P = 0.47). Among LoFs, the strongest burden was observed for INIT (OR = 2.16, P = 0.0097) and premature stop variants predicted to cause non-sense-mediated decay in the majority of transcripts (NMD) (OR = 1.98, P = 0.02). Notably, this increased burden of NMD, INIT and splice variants was more pronounced in a set of 1397 innate immune genes (OR = 4.55, P = 0.0043). Further comparison to additional exomes indicates that the difference in LoF burden originated both from the AD and centenarian sample. In summary, we observed an overall increased burden of rare LoFs in AD subjects as compared to centenarians, and this enrichment is more pronounced for innate immune genes.
Collapse
Affiliation(s)
- Yun Freudenberg-Hua
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030, USA .,Division of Geriatric Psychiatry, Zucker Hillside Hospital, Northwell Health, Glen Oaks, NY 11004, USA
| | - Wentian Li
- Robert S Boas Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030, USA
| | | | | | - Vanessa Cortes
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030, USA
| | - Danny Ben-Avraham
- Institute for Aging Research and the Diabetes Research Center, Department of Medicine and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jeremy Koppel
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030, USA
| | - Blaine Greenwald
- Division of Geriatric Psychiatry, Zucker Hillside Hospital, Northwell Health, Glen Oaks, NY 11004, USA
| | | | | | | | - Nir Barzilai
- Institute for Aging Research and the Diabetes Research Center, Department of Medicine and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jan Freudenberg
- Robert S Boas Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030, USA.,The Regeneron Genetics Center, Tarrytown, NY 10591, USA and
| | - Gil Atzmon
- Institute for Aging Research and the Diabetes Research Center, Department of Medicine and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Faculty of Natural Sciences, University of Haifa, Haifa 31905, Israel
| | - Peter Davies
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030, USA
| |
Collapse
|
39
|
|
40
|
Garatachea N, Marín PJ, Santos-Lozano A, Sanchis-Gomar F, Emanuele E, Lucia A. The ApoE gene is related with exceptional longevity: a systematic review and meta-analysis. Rejuvenation Res 2016; 18:3-13. [PMID: 25385258 DOI: 10.1089/rej.2014.1605] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The objective of this meta-analysis was to determine the association of the apolipoprotein E (ApoE) gene with exceptional longevity (EL) (i.e., reaching 100+ years) by identifying possible unequal distribution of alleles/genotypes in the common variants ε2, ε3, and ε4 among centenarians and younger population. The association of ApoE with EL was analyzed in a total of 2776 centenarians (cases) and 11,941 younger controls (from 13 case-control studies) using the chi-squared test with the Yates correction. We conducted combined and separate analyses for all ethnic groups studied in the literature (Caucasian and Asian). The main result for all ethnic groups combined was that the likelihood of reaching EL was negatively associated with ε4 allele carriage [pooled odds ratio (OR)=0.43; 95% confidence interval (CI) 0.36, 0.50; p<0.001] and with ε4/ε4 (OR=0.18; 95% CI 0.08, 0.39; p<0.001), ε3/ε4 (OR=0.44; 95% CI 0.37, 0.53; p<0.001) and ε2/ε4 genotypes (OR=0.48; 95% CI 0.31, 0.74; p<0.001). In contrast, the ε2/ε3 genotype was positively associated with EL (OR=1.35; 95% CI 1.06, 1.72; p=0.017). When compared with the ε3 allele, the ε2 allele was not associated with increased odds of EL (OR=1.08; 95% CI 0.77, 1.50, p=0.660). The present meta-analysis confirms that, besides its previously documented influence on Alzheimer's and cardiovascular disease risk, the ApoE gene is associated with the likelihood of reaching EL.
Collapse
Affiliation(s)
- Nuria Garatachea
- 1 Faculty of Health and Sport Science, University of Zaragoza , Huesca, Spain
| | | | | | | | | | | |
Collapse
|
41
|
Butkiewicz M, Bush WS. In Silico Functional Annotation of Genomic Variation. CURRENT PROTOCOLS IN HUMAN GENETICS 2016; 88:6.15.1-6.15.17. [PMID: 26724722 PMCID: PMC4722816 DOI: 10.1002/0471142905.hg0615s88] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This unit describes the concepts and practical techniques for annotating genomic variants in the human genome to estimate their functional significance. With the rapid increase of available whole exome and whole genome sequencing information for human studies, annotation techniques have become progressively more important for highlighting and prioritizing nucleotide variants and their potential impact on genes and other genetic constructs. Here, we present an overview of different types of variant annotation approaches and elaborate on their foundations, assumptions, and the downstream consequences of their use. Computational approaches and tools to assign annotations and to identify variants are reviewed. Further, the general philosophy of assigning potential function to a genetic change within the biological context of a disease is discussed.
Collapse
Affiliation(s)
- Mariusz Butkiewicz
- Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio
| | - William S Bush
- Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
42
|
Fortney K, Dobriban E, Garagnani P, Pirazzini C, Monti D, Mari D, Atzmon G, Barzilai N, Franceschi C, Owen AB, Kim SK. Genome-Wide Scan Informed by Age-Related Disease Identifies Loci for Exceptional Human Longevity. PLoS Genet 2015; 11:e1005728. [PMID: 26677855 PMCID: PMC4683064 DOI: 10.1371/journal.pgen.1005728] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Accepted: 11/16/2015] [Indexed: 11/20/2022] Open
Abstract
We developed a new statistical framework to find genetic variants associated with extreme longevity. The method, informed GWAS (iGWAS), takes advantage of knowledge from large studies of age-related disease in order to narrow the search for SNPs associated with longevity. To gain support for our approach, we first show there is an overlap between loci involved in disease and loci associated with extreme longevity. These results indicate that several disease variants may be depleted in centenarians versus the general population. Next, we used iGWAS to harness information from 14 meta-analyses of disease and trait GWAS to identify longevity loci in two studies of long-lived humans. In a standard GWAS analysis, only one locus in these studies is significant (APOE/TOMM40) when controlling the false discovery rate (FDR) at 10%. With iGWAS, we identify eight genetic loci to associate significantly with exceptional human longevity at FDR < 10%. We followed up the eight lead SNPs in independent cohorts, and found replication evidence of four loci and suggestive evidence for one more with exceptional longevity. The loci that replicated (FDR < 5%) included APOE/TOMM40 (associated with Alzheimer’s disease), CDKN2B/ANRIL (implicated in the regulation of cellular senescence), ABO (tags the O blood group), and SH2B3/ATXN2 (a signaling gene that extends lifespan in Drosophila and a gene involved in neurological disease). Our results implicate new loci in longevity and reveal a genetic overlap between longevity and age-related diseases and traits, including coronary artery disease and Alzheimer’s disease. iGWAS provides a new analytical strategy for uncovering SNPs that influence extreme longevity, and can be applied more broadly to boost power in other studies of complex phenotypes. Longevity is a complex phenotype, and few genetic variants that affect lifespan have been identified. However, aging and disease are closely related, and a great deal is known about the genetic basis of disease risk. Here, we show using genome-wide association studies (GWAS) of longevity and disease that there is an overlap between loci involved in longevity and loci involved in several diseases, such as Alzheimer’s disease and coronary artery disease. We then develop a new statistical framework to find genetic variants associated with extreme longevity. The method, informed GWAS (iGWAS), takes advantage of knowledge from 14 large studies of disease and disease-related traits in order to narrow the search for SNPs associated with longevity. Using iGWAS, we found eight SNPs that are significant in our discovery cohorts, and we were able to validate four of these in replication studies of long-lived subjects. Our results implicate new loci in longevity and reveal a genetic overlap between longevity and age-related diseases and traits. Beyond the study of human longevity, iGWAS can be applied to boost statistical power in any GWAS of a target phenotype by using larger GWAS of genetically-related conditions.
Collapse
Affiliation(s)
- Kristen Fortney
- Department of Developmental Biology, Stanford University, Stanford, California, United States of America
- Department of Genetics, Stanford University, Stanford, California, United States of America
| | - Edgar Dobriban
- Department of Statistics, Stanford University, Stanford, California, United States of America
| | - Paolo Garagnani
- Department of Experimental, Diagnostic and Specialty Medicine Experimental Pathology, University of Bologna, Bologna, Italy
- Center for Applied Biomedical Research, St. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Chiara Pirazzini
- Department of Experimental, Diagnostic and Specialty Medicine Experimental Pathology, University of Bologna, Bologna, Italy
- Interdepartmental Centre "L. Galvani" CIG, University of Bologna, Bologna, Italy
| | - Daniela Monti
- Department of Clinical, Experimental and Biomedical Sciences, University of Florence, Florence, Italy
| | - Daniela Mari
- Department of Medical Sciences, University of Milan, Milan, Italy
- Geriatric Unit, IRCCS Ca' Grande Foundation, Maggiore Policlinico Hospital, Milan, Italy
| | - Gil Atzmon
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Nir Barzilai
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Claudio Franceschi
- Department of Experimental, Diagnostic and Specialty Medicine Experimental Pathology, University of Bologna, Bologna, Italy
- IRCCS, Institute of Neurological Sciences of Bologna, Bologna, Italy
| | - Art B. Owen
- Department of Statistics, Stanford University, Stanford, California, United States of America
| | - Stuart K. Kim
- Department of Developmental Biology, Stanford University, Stanford, California, United States of America
- Department of Genetics, Stanford University, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
43
|
Milman S, Barzilai N. Dissecting the Mechanisms Underlying Unusually Successful Human Health Span and Life Span. Cold Spring Harb Perspect Med 2015; 6:a025098. [PMID: 26637439 DOI: 10.1101/cshperspect.a025098] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Humans age at different rates and families with exceptional survival provide the opportunity to understand why some people age slower than others. Unique features exhibited by centenarians include a family history of longevity, compression of morbidity with resultant extension of health span, and biomarkers such as low-circulating insulin-like growth factor 1 (IGF-1) and elevated high-density lipoprotein (HDL) cholesterol levels. Given the rarity of the centenarian phenotype, it has not been surprising that the use of discovery methods that relied on common population single nucleotide polymorphisms (SNPs) to unlock the genetic determinants of exceptional longevity have not yielded significant results. Conversely, gene sequencing has resulted in discoveries of functional gene variants that support several of the centenarian phenotypes. These discoveries have led to the strategic developments of drugs that may delay aging and prolong health span.
Collapse
Affiliation(s)
- Sofiya Milman
- Department of Medicine, Division of Endocrinology, Albert Einstein College of Medicine, New York, New York 10461 Institute for Aging Research, Albert Einstein College of Medicine, New York, New York 10461
| | - Nir Barzilai
- Department of Medicine, Division of Endocrinology, Albert Einstein College of Medicine, New York, New York 10461 Institute for Aging Research, Albert Einstein College of Medicine, New York, New York 10461 Department of Genetics, Albert Einstein College of Medicine, Bronx, New York 10461
| |
Collapse
|
44
|
Horvath S, Pirazzini C, Bacalini MG, Gentilini D, Di Blasio AM, Delledonne M, Mari D, Arosio B, Monti D, Passarino G, De Rango F, D'Aquila P, Giuliani C, Marasco E, Collino S, Descombes P, Garagnani P, Franceschi C. Decreased epigenetic age of PBMCs from Italian semi-supercentenarians and their offspring. Aging (Albany NY) 2015; 7:1159-70. [PMID: 26678252 PMCID: PMC4712339 DOI: 10.18632/aging.100861] [Citation(s) in RCA: 212] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Given the dramatic increase in ageing populations, it is of great importance to understand the genetic and molecular determinants of healthy ageing and longevity. Semi-supercentenarians (subjects who reached an age of 105-109 years) arguably represent the gold standard of successful human ageing because they managed to avoid or postpone the onset of major age-related diseases. Relatively few studies have looked at epigenetic determinants of extreme longevity in humans. Here we test whether families with extreme longevity are epigenetically distinct from controls according to an epigenetic biomarker of ageing which is known as "epigenetic clock". We analyze the DNA methylation levels of peripheral blood mononuclear cells (PBMCs) from Italian families constituted of 82 semi-supercentenarians (mean age: 105.6 ± 1.6 years), 63 semi-supercentenarians' offspring (mean age: 71.8 ± 7.8 years), and 47 age-matched controls (mean age: 69.8 ± 7.2 years). We demonstrate that the offspring of semi-supercentenarians have a lower epigenetic age than age-matched controls (age difference=5.1 years, p=0.00043) and that centenarians are younger (8.6 years) than expected based on their chronological age. By contrast, no significant difference could be observed for estimated blood cell counts (such as naïve or exhausted cytotoxic T cells or helper T cells). Future studies will be needed to replicate these findings in different populations and to extend them to other tissues. Overall, our results suggest that epigenetic processes might play a role in extreme longevity and healthy human ageing.
Collapse
Affiliation(s)
- Steve Horvath
- Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Biostatistics, School of Public Health, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Chiara Pirazzini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40126 Bologna, Italy
- Interdepartmental Center "L. Galvani", University of Bologna, 40126 Bologna, Italy
| | - Maria Giulia Bacalini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40126 Bologna, Italy
- Interdepartmental Center "L. Galvani", University of Bologna, 40126 Bologna, Italy
- Personal Genomics S.r.l., 37134 Verona, Italy
| | - Davide Gentilini
- Istituto Auxologico Italiano IRCCS, Cusano Milanino, 20095 Milan, Italy
| | | | - Massimo Delledonne
- Personal Genomics S.r.l., 37134 Verona, Italy
- Functional Genomics Center, Department of Biotechnology, University of Verona, 37134 Verona, Italy
| | - Daniela Mari
- Geriatric Unit, Department of Medical Sciences and Community Health, University of Milan, 20122 Milan, Italy
- Geriatric Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Beatrice Arosio
- Geriatric Unit, Department of Medical Sciences and Community Health, University of Milan, 20122 Milan, Italy
- Geriatric Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Daniela Monti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy
| | | | | | - Patrizia D'Aquila
- Department of Cell Biology, University of Calabria, 87036 Rende, Italy
| | - Cristina Giuliani
- Department of Biological, Geological and Environmental Sciences, Laboratory of Molecular Anthropology and Centre for Genome Biology, University of Bologna, 40126 Bologna, Italy
| | - Elena Marasco
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40126 Bologna, Italy
- Interdepartmental Center "L. Galvani", University of Bologna, 40126 Bologna, Italy
| | - Sebastiano Collino
- Molecular Biomarkers, Nestlé Institute of Health Sciences SA, 1015 Lausanne, Switzerland
| | - Patrick Descombes
- Functional Genomics, Nestlé Institute of Health Sciences SA, 1015 Lausanne, Switzerland
| | - Paolo Garagnani
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40126 Bologna, Italy
- Interdepartmental Center "L. Galvani", University of Bologna, 40126 Bologna, Italy
- CRBA, Center for Applied Biomedical Research, St. Orsola-Malpighi University Hospital, 40138 Bologna, Italy
| | - Claudio Franceschi
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40126 Bologna, Italy
- Interdepartmental Center "L. Galvani", University of Bologna, 40126 Bologna, Italy
- CNR, Institute of Organic Synthesis and Photoreactivity (ISOF), 40129 Bologna, Italy
- IRCCS, Institute of Neurological Sciences of Bologna, 40139 Bologna, Italy
| |
Collapse
|
45
|
Puzzling role of genetic risk factors in human longevity: "risk alleles" as pro-longevity variants. Biogerontology 2015; 17:109-27. [PMID: 26306600 PMCID: PMC4724477 DOI: 10.1007/s10522-015-9600-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 08/19/2015] [Indexed: 02/07/2023]
Abstract
Complex diseases are major contributors to human mortality in old age. Paradoxically, many genetic variants that have been associated with increased risks of such diseases are found in genomes of long-lived people, and do not seem to compromise longevity. Here we argue that trade-off-like and conditional effects of genes can play central role in this phenomenon and in determining longevity. Such effects may occur as result of: (i) antagonistic influence of gene on the development of different health disorders; (ii) change in the effect of gene on vulnerability to death with age (especially, from “bad” to “good”); (iii) gene–gene interaction; and (iv) gene–environment interaction, among other factors. A review of current knowledge provides many examples of genetic factors that may increase the risk of one disease but reduce chances of developing another serious health condition, or improve survival from it. Factors that may increase risk of a major disease but attenuate manifestation of physical senescence are also discussed. Overall, available evidence suggests that the influence of a genetic variant on longevity may be negative, neutral or positive, depending on a delicate balance of the detrimental and beneficial effects of such variant on multiple health and aging related traits. This balance may change with age, internal and external environments, and depend on genetic surrounding. We conclude that trade-off-like and conditional genetic effects are very common and may result in situations when a disease “risk allele” can also be a pro-longevity variant, depending on context. We emphasize importance of considering such effects in both aging research and disease prevention.
Collapse
|
46
|
Castillo-Quan JI, Kinghorn KJ, Bjedov I. Genetics and pharmacology of longevity: the road to therapeutics for healthy aging. ADVANCES IN GENETICS 2015; 90:1-101. [PMID: 26296933 DOI: 10.1016/bs.adgen.2015.06.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Aging can be defined as the progressive decline in tissue and organismal function and the ability to respond to stress that occurs in association with homeostatic failure and the accumulation of molecular damage. Aging is the biggest risk factor for human disease and results in a wide range of aging pathologies. Although we do not completely understand the underlying molecular basis that drives the aging process, we have gained exceptional insights into the plasticity of life span and healthspan from the use of model organisms such as the worm Caenorhabditis elegans and the fruit fly Drosophila melanogaster. Single-gene mutations in key cellular pathways that regulate environmental sensing, and the response to stress, have been identified that prolong life span across evolution from yeast to mammals. These genetic manipulations also correlate with a delay in the onset of tissue and organismal dysfunction. While the molecular genetics of aging will remain a prosperous and attractive area of research in biogerontology, we are moving towards an era defined by the search for therapeutic drugs that promote healthy aging. Translational biogerontology will require incorporation of both therapeutic and pharmacological concepts. The use of model organisms will remain central to the quest for drug discovery, but as we uncover molecular processes regulated by repurposed drugs and polypharmacy, studies of pharmacodynamics and pharmacokinetics, drug-drug interactions, drug toxicity, and therapeutic index will slowly become more prevalent in aging research. As we move from genetics to pharmacology and therapeutics, studies will not only require demonstration of life span extension and an underlying molecular mechanism, but also the translational relevance for human health and disease prevention.
Collapse
Affiliation(s)
- Jorge Iván Castillo-Quan
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK; Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Kerri J Kinghorn
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK; Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Ivana Bjedov
- Cancer Institute, University College London, London, UK
| |
Collapse
|
47
|
Stevenson M, Bae H, Schupf N, Andersen S, Zhang Q, Perls T, Sebastiani P. Burden of disease variants in participants of the Long Life Family Study. Aging (Albany NY) 2015; 7:123-32. [PMID: 25664523 PMCID: PMC4359694 DOI: 10.18632/aging.100724] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Case control studies of nonagenarians and centenarians provide evidence that long-lived individuals do not differ in the rate of disease associated variants compared to population controls. These results suggest that an enrichment of novel protective variants, rather than a lack of disease associated variants, determine the genetic predisposition to exceptionally long lives. Using data from the Long Life Family Study (LLFS), we sought to replicate these findings and extend them to include a larger number of disease-specific risk alleles. To accomplish this goal, we built a genetic risk score for each of four age-related disease groups: Alzheimer's disease, cardiovascular disease and stroke, type 2 diabetes, and various cancers and compared the distribution of these scores between older participants of the LLFS, their offspring and their spouses. The analyses showed no significant differences in distribution of the genetic risk scores for cardiovascular disease and stroke, type 2 diabetes, or cancer between the groups, while participants of the LLFS appeared to carry an average 1% fewer risk alleles for Alzheimer's disease compared to spousal controls and, while the difference may not be clinically relevant, it was statistically significant. However, the statistical significance between familial longevity and the Alzheimer's disease genetic risk score was lost when a more stringent linkage disequilibrium threshold was imposed to select independent genetic variants.
Collapse
Affiliation(s)
- Meredith Stevenson
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Harold Bae
- College of Public Health and Human Sciences, Oregon State University, OR 97331, USA
| | - Nicole Schupf
- Department of Neurology, Columbia UNiversity, New York City, NY 10027, USA
| | - Stacy Andersen
- Section of Geriatrics, Department of Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA 02118, USA
| | - Qunyuan Zhang
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Thomas Perls
- Section of Geriatrics, Department of Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA 02118, USA
| | - Paola Sebastiani
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| |
Collapse
|
48
|
Abstract
![]()
Whole human genome sequencing of
individuals is becoming rapid
and inexpensive, enabling new strategies for using personal genome
information to help diagnose, treat, and even prevent human disorders
for which genetic variations are causative or are known to be risk
factors. Many of the exploding number of newly discovered genetic
variations alter the structure, function, dynamics, stability, and/or
interactions of specific proteins and RNA molecules. Accordingly,
there are a host of opportunities for biochemists and biophysicists
to participate in (1) developing tools to allow accurate and sometimes
medically actionable assessment of the potential pathogenicity of
individual variations and (2) establishing the mechanistic linkage
between pathogenic variations and their physiological consequences,
providing a rational basis for treatment or preventive care. In this
review, we provide an overview of these opportunities and their associated
challenges in light of the current status of genomic science and personalized
medicine, the latter often termed precision medicine.
Collapse
Affiliation(s)
- Brett M Kroncke
- †Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States.,‡Center for Structural Biology, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Carlos G Vanoye
- §Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Jens Meiler
- ‡Center for Structural Biology, Vanderbilt University, Nashville, Tennessee 37232, United States.,∥Departments of Chemistry, Pharmacology, and Bioinformatics, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Alfred L George
- §Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Charles R Sanders
- †Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States.,‡Center for Structural Biology, Vanderbilt University, Nashville, Tennessee 37232, United States
| |
Collapse
|
49
|
Yashin AI, Wu D, Arbeeva LS, Arbeev KG, Kulminski AM, Akushevich I, Kovtun M, Culminskaya I, Stallard E, Li M, Ukraintseva SV. Genetics of aging, health, and survival: dynamic regulation of human longevity related traits. Front Genet 2015; 6:122. [PMID: 25918517 PMCID: PMC4394697 DOI: 10.3389/fgene.2015.00122] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 03/16/2015] [Indexed: 11/13/2022] Open
Abstract
Background: The roles of genetic factors in human longevity would be better understood if one can use more efficient methods in genetic analyses and investigate pleiotropic effects of genetic variants on aging and health related traits. Data and methods: We used EMMAX software with modified correction for population stratification to perform genome wide association studies (GWAS) of female lifespan from the original FHS cohort. The male data from the original FHS cohort and male and female data combined from the offspring FHS cohort were used to confirm findings. We evaluated pleiotropic effects of selected genetic variants as well as gene-smoking interactions on health and aging related traits. Then we reviewed current knowledge on functional properties of genes related to detected variants. Results: The eight SNPs with genome-wide significant variants were negatively associated with lifespan in both males and females. After additional QC, two of these variants were selected for further analyses of their associations with major diseases (cancer and CHD) and physiological aging changes. Gene-smoking interactions contributed to these effects. Genes closest to detected variants appear to be involved in similar biological processes and health disorders, as those found in other studies of aging and longevity e.g., in cancer and neurodegeneration. Conclusions: The impact of genes on longevity may involve trade-off-like effects on different health traits. Genes that influence lifespan represent various molecular functions but may be involved in similar biological processes and health disorders, which could contribute to genetic heterogeneity of longevity and the lack of replication in genetic association studies.
Collapse
Affiliation(s)
- Anatoliy I Yashin
- Biodemography of Aging Research Unit, Center for Population Health and Aging, Social Science Research Institute, Duke University Durham, NC, USA
| | - Deqing Wu
- Biodemography of Aging Research Unit, Center for Population Health and Aging, Social Science Research Institute, Duke University Durham, NC, USA
| | - Liubov S Arbeeva
- Biodemography of Aging Research Unit, Center for Population Health and Aging, Social Science Research Institute, Duke University Durham, NC, USA
| | - Konstantin G Arbeev
- Biodemography of Aging Research Unit, Center for Population Health and Aging, Social Science Research Institute, Duke University Durham, NC, USA
| | - Alexander M Kulminski
- Biodemography of Aging Research Unit, Center for Population Health and Aging, Social Science Research Institute, Duke University Durham, NC, USA
| | - Igor Akushevich
- Biodemography of Aging Research Unit, Center for Population Health and Aging, Social Science Research Institute, Duke University Durham, NC, USA
| | - Mikhail Kovtun
- Biodemography of Aging Research Unit, Center for Population Health and Aging, Social Science Research Institute, Duke University Durham, NC, USA ; Integrative Genomic Analysis Shared Resource, Duke Center for Genomic and Computational Biology, Duke University Durham, NC, USA
| | - Irina Culminskaya
- Biodemography of Aging Research Unit, Center for Population Health and Aging, Social Science Research Institute, Duke University Durham, NC, USA
| | - Eric Stallard
- Biodemography of Aging Research Unit, Center for Population Health and Aging, Social Science Research Institute, Duke University Durham, NC, USA
| | - Miaozhu Li
- Biodemography of Aging Research Unit, Center for Population Health and Aging, Social Science Research Institute, Duke University Durham, NC, USA
| | - Svetlana V Ukraintseva
- Biodemography of Aging Research Unit, Center for Population Health and Aging, Social Science Research Institute, Duke University Durham, NC, USA
| |
Collapse
|
50
|
Toledo RA, Hatakana R, Lourenço DM, Lindsey SC, Camacho CP, Almeida M, Lima JV, Sekiya T, Garralda E, Naslavsky MS, Yamamoto GL, Lazar M, Meirelles O, Sobreira TJP, Lebrao ML, Duarte YAO, Blangero J, Zatz M, Cerutti JM, Maciel RMB, Toledo SPA. Comprehensive assessment of the disputed RET Y791F variant shows no association with medullary thyroid carcinoma susceptibility. Endocr Relat Cancer 2015; 22:65-76. [PMID: 25425582 PMCID: PMC4289937 DOI: 10.1530/erc-14-0491] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Accurate interpretation of germline mutations of the rearranged during transfection (RET) proto-oncogene is vital for the proper recommendation of preventive thyroidectomy in medullary thyroid carcinoma (MTC)-prone carriers. To gain information regarding the most disputed variant of RET, ATA-A Y791F, we sequenced blood DNA samples from a cohort of 2904 cancer-free elderly individuals (1261 via Sanger sequencing and 1643 via whole-exome/genome sequencing). We also accessed the exome sequences of an additional 8069 individuals from non-cancer-related laboratories and public databanks as well as genetic results from the Catalogue of Somatic Mutations in Cancer (COSMIC) project. The mean allelic frequency observed in the controls was 0.0031, with higher occurrences in Central European populations (0.006/0.008). The prevalence of RET Y791F in the control databases was extremely high compared with the 40 known RET pathogenic mutations (P=0.00003), while no somatic occurrence has been reported in tumours. In this study, we report new, unrelated Brazilian individuals with germline RET Y791F-only: two tumour-free elderly controls; two individuals with sporadic MTC whose Y791F-carrying relatives did not show any evidence of tumours; and a 74-year-old phaeochromocytoma patient without MTC. Furthermore, we showed that the co-occurrence of Y791F with the strong RET C634Y mutation explains the aggressive MTC phenotypes observed in a large affected family that was initially reported as Y791F-only. Our literature review revealed that limited analyses have led to the misclassification of RET Y791F as a probable pathogenic variant and, consequently, to the occurrence of unnecessary thyroidectomies. The current study will have a substantial clinical influence, as it reveals, in a comprehensive manner, that RET Y791F only shows no association with MTC susceptibility.
Collapse
Affiliation(s)
- Rodrigo A Toledo
- Endocrine Genetics Unit (Laboratório de Investigação Médica/LIM-25) of Hospital das ClínicasUniversity of São Paulo School of Medicine, São Paulo, São Paulo 05403-010, BrazilNursing SchoolSchool of Public HealthHuman Genome Research CenterUniversity of São Paulo, São Paulo, São Paulo, BrazilDivision of GeneticsGenetic Bases of Thyroid Tumors Laboratory, Department of Morphology and GeneticsDivision of EndocrinologyLaboratory of Molecular and Translational Endocrinology, Department of Medicine, Federal University of São Paulo, São Paulo, São Paulo, BrazilBrazilian National Laboratory of BiosciencesCampinas, São Paulo, BrazilCentro Integral Oncológico Clara CampalHospital Universitário Sanchinarro, Madrid, SpainDepartment of GeneticsTexas Biomedical Research Institute, AT&T Genomic Computing Center, San Antonio, Texas, USAEndocrinology DivisionSanta Casa Hospital, São Paulo, São Paulo, BrazilLaboratory of Epidemiology and Population SciencesNational Institute on Aging, Bethesda, Maryland, USA
| | - Roxanne Hatakana
- Endocrine Genetics Unit (Laboratório de Investigação Médica/LIM-25) of Hospital das ClínicasUniversity of São Paulo School of Medicine, São Paulo, São Paulo 05403-010, BrazilNursing SchoolSchool of Public HealthHuman Genome Research CenterUniversity of São Paulo, São Paulo, São Paulo, BrazilDivision of GeneticsGenetic Bases of Thyroid Tumors Laboratory, Department of Morphology and GeneticsDivision of EndocrinologyLaboratory of Molecular and Translational Endocrinology, Department of Medicine, Federal University of São Paulo, São Paulo, São Paulo, BrazilBrazilian National Laboratory of BiosciencesCampinas, São Paulo, BrazilCentro Integral Oncológico Clara CampalHospital Universitário Sanchinarro, Madrid, SpainDepartment of GeneticsTexas Biomedical Research Institute, AT&T Genomic Computing Center, San Antonio, Texas, USAEndocrinology DivisionSanta Casa Hospital, São Paulo, São Paulo, BrazilLaboratory of Epidemiology and Population SciencesNational Institute on Aging, Bethesda, Maryland, USA
| | - Delmar M Lourenço
- Endocrine Genetics Unit (Laboratório de Investigação Médica/LIM-25) of Hospital das ClínicasUniversity of São Paulo School of Medicine, São Paulo, São Paulo 05403-010, BrazilNursing SchoolSchool of Public HealthHuman Genome Research CenterUniversity of São Paulo, São Paulo, São Paulo, BrazilDivision of GeneticsGenetic Bases of Thyroid Tumors Laboratory, Department of Morphology and GeneticsDivision of EndocrinologyLaboratory of Molecular and Translational Endocrinology, Department of Medicine, Federal University of São Paulo, São Paulo, São Paulo, BrazilBrazilian National Laboratory of BiosciencesCampinas, São Paulo, BrazilCentro Integral Oncológico Clara CampalHospital Universitário Sanchinarro, Madrid, SpainDepartment of GeneticsTexas Biomedical Research Institute, AT&T Genomic Computing Center, San Antonio, Texas, USAEndocrinology DivisionSanta Casa Hospital, São Paulo, São Paulo, BrazilLaboratory of Epidemiology and Population SciencesNational Institute on Aging, Bethesda, Maryland, USA
| | - Susan C Lindsey
- Endocrine Genetics Unit (Laboratório de Investigação Médica/LIM-25) of Hospital das ClínicasUniversity of São Paulo School of Medicine, São Paulo, São Paulo 05403-010, BrazilNursing SchoolSchool of Public HealthHuman Genome Research CenterUniversity of São Paulo, São Paulo, São Paulo, BrazilDivision of GeneticsGenetic Bases of Thyroid Tumors Laboratory, Department of Morphology and GeneticsDivision of EndocrinologyLaboratory of Molecular and Translational Endocrinology, Department of Medicine, Federal University of São Paulo, São Paulo, São Paulo, BrazilBrazilian National Laboratory of BiosciencesCampinas, São Paulo, BrazilCentro Integral Oncológico Clara CampalHospital Universitário Sanchinarro, Madrid, SpainDepartment of GeneticsTexas Biomedical Research Institute, AT&T Genomic Computing Center, San Antonio, Texas, USAEndocrinology DivisionSanta Casa Hospital, São Paulo, São Paulo, BrazilLaboratory of Epidemiology and Population SciencesNational Institute on Aging, Bethesda, Maryland, USA
| | - Cleber P Camacho
- Endocrine Genetics Unit (Laboratório de Investigação Médica/LIM-25) of Hospital das ClínicasUniversity of São Paulo School of Medicine, São Paulo, São Paulo 05403-010, BrazilNursing SchoolSchool of Public HealthHuman Genome Research CenterUniversity of São Paulo, São Paulo, São Paulo, BrazilDivision of GeneticsGenetic Bases of Thyroid Tumors Laboratory, Department of Morphology and GeneticsDivision of EndocrinologyLaboratory of Molecular and Translational Endocrinology, Department of Medicine, Federal University of São Paulo, São Paulo, São Paulo, BrazilBrazilian National Laboratory of BiosciencesCampinas, São Paulo, BrazilCentro Integral Oncológico Clara CampalHospital Universitário Sanchinarro, Madrid, SpainDepartment of GeneticsTexas Biomedical Research Institute, AT&T Genomic Computing Center, San Antonio, Texas, USAEndocrinology DivisionSanta Casa Hospital, São Paulo, São Paulo, BrazilLaboratory of Epidemiology and Population SciencesNational Institute on Aging, Bethesda, Maryland, USA
| | - Marcio Almeida
- Endocrine Genetics Unit (Laboratório de Investigação Médica/LIM-25) of Hospital das ClínicasUniversity of São Paulo School of Medicine, São Paulo, São Paulo 05403-010, BrazilNursing SchoolSchool of Public HealthHuman Genome Research CenterUniversity of São Paulo, São Paulo, São Paulo, BrazilDivision of GeneticsGenetic Bases of Thyroid Tumors Laboratory, Department of Morphology and GeneticsDivision of EndocrinologyLaboratory of Molecular and Translational Endocrinology, Department of Medicine, Federal University of São Paulo, São Paulo, São Paulo, BrazilBrazilian National Laboratory of BiosciencesCampinas, São Paulo, BrazilCentro Integral Oncológico Clara CampalHospital Universitário Sanchinarro, Madrid, SpainDepartment of GeneticsTexas Biomedical Research Institute, AT&T Genomic Computing Center, San Antonio, Texas, USAEndocrinology DivisionSanta Casa Hospital, São Paulo, São Paulo, BrazilLaboratory of Epidemiology and Population SciencesNational Institute on Aging, Bethesda, Maryland, USA
| | - José V Lima
- Endocrine Genetics Unit (Laboratório de Investigação Médica/LIM-25) of Hospital das ClínicasUniversity of São Paulo School of Medicine, São Paulo, São Paulo 05403-010, BrazilNursing SchoolSchool of Public HealthHuman Genome Research CenterUniversity of São Paulo, São Paulo, São Paulo, BrazilDivision of GeneticsGenetic Bases of Thyroid Tumors Laboratory, Department of Morphology and GeneticsDivision of EndocrinologyLaboratory of Molecular and Translational Endocrinology, Department of Medicine, Federal University of São Paulo, São Paulo, São Paulo, BrazilBrazilian National Laboratory of BiosciencesCampinas, São Paulo, BrazilCentro Integral Oncológico Clara CampalHospital Universitário Sanchinarro, Madrid, SpainDepartment of GeneticsTexas Biomedical Research Institute, AT&T Genomic Computing Center, San Antonio, Texas, USAEndocrinology DivisionSanta Casa Hospital, São Paulo, São Paulo, BrazilLaboratory of Epidemiology and Population SciencesNational Institute on Aging, Bethesda, Maryland, USA
| | - Tomoko Sekiya
- Endocrine Genetics Unit (Laboratório de Investigação Médica/LIM-25) of Hospital das ClínicasUniversity of São Paulo School of Medicine, São Paulo, São Paulo 05403-010, BrazilNursing SchoolSchool of Public HealthHuman Genome Research CenterUniversity of São Paulo, São Paulo, São Paulo, BrazilDivision of GeneticsGenetic Bases of Thyroid Tumors Laboratory, Department of Morphology and GeneticsDivision of EndocrinologyLaboratory of Molecular and Translational Endocrinology, Department of Medicine, Federal University of São Paulo, São Paulo, São Paulo, BrazilBrazilian National Laboratory of BiosciencesCampinas, São Paulo, BrazilCentro Integral Oncológico Clara CampalHospital Universitário Sanchinarro, Madrid, SpainDepartment of GeneticsTexas Biomedical Research Institute, AT&T Genomic Computing Center, San Antonio, Texas, USAEndocrinology DivisionSanta Casa Hospital, São Paulo, São Paulo, BrazilLaboratory of Epidemiology and Population SciencesNational Institute on Aging, Bethesda, Maryland, USA
| | - Elena Garralda
- Endocrine Genetics Unit (Laboratório de Investigação Médica/LIM-25) of Hospital das ClínicasUniversity of São Paulo School of Medicine, São Paulo, São Paulo 05403-010, BrazilNursing SchoolSchool of Public HealthHuman Genome Research CenterUniversity of São Paulo, São Paulo, São Paulo, BrazilDivision of GeneticsGenetic Bases of Thyroid Tumors Laboratory, Department of Morphology and GeneticsDivision of EndocrinologyLaboratory of Molecular and Translational Endocrinology, Department of Medicine, Federal University of São Paulo, São Paulo, São Paulo, BrazilBrazilian National Laboratory of BiosciencesCampinas, São Paulo, BrazilCentro Integral Oncológico Clara CampalHospital Universitário Sanchinarro, Madrid, SpainDepartment of GeneticsTexas Biomedical Research Institute, AT&T Genomic Computing Center, San Antonio, Texas, USAEndocrinology DivisionSanta Casa Hospital, São Paulo, São Paulo, BrazilLaboratory of Epidemiology and Population SciencesNational Institute on Aging, Bethesda, Maryland, USA
| | - Michel S Naslavsky
- Endocrine Genetics Unit (Laboratório de Investigação Médica/LIM-25) of Hospital das ClínicasUniversity of São Paulo School of Medicine, São Paulo, São Paulo 05403-010, BrazilNursing SchoolSchool of Public HealthHuman Genome Research CenterUniversity of São Paulo, São Paulo, São Paulo, BrazilDivision of GeneticsGenetic Bases of Thyroid Tumors Laboratory, Department of Morphology and GeneticsDivision of EndocrinologyLaboratory of Molecular and Translational Endocrinology, Department of Medicine, Federal University of São Paulo, São Paulo, São Paulo, BrazilBrazilian National Laboratory of BiosciencesCampinas, São Paulo, BrazilCentro Integral Oncológico Clara CampalHospital Universitário Sanchinarro, Madrid, SpainDepartment of GeneticsTexas Biomedical Research Institute, AT&T Genomic Computing Center, San Antonio, Texas, USAEndocrinology DivisionSanta Casa Hospital, São Paulo, São Paulo, BrazilLaboratory of Epidemiology and Population SciencesNational Institute on Aging, Bethesda, Maryland, USA
| | - Guilherme L Yamamoto
- Endocrine Genetics Unit (Laboratório de Investigação Médica/LIM-25) of Hospital das ClínicasUniversity of São Paulo School of Medicine, São Paulo, São Paulo 05403-010, BrazilNursing SchoolSchool of Public HealthHuman Genome Research CenterUniversity of São Paulo, São Paulo, São Paulo, BrazilDivision of GeneticsGenetic Bases of Thyroid Tumors Laboratory, Department of Morphology and GeneticsDivision of EndocrinologyLaboratory of Molecular and Translational Endocrinology, Department of Medicine, Federal University of São Paulo, São Paulo, São Paulo, BrazilBrazilian National Laboratory of BiosciencesCampinas, São Paulo, BrazilCentro Integral Oncológico Clara CampalHospital Universitário Sanchinarro, Madrid, SpainDepartment of GeneticsTexas Biomedical Research Institute, AT&T Genomic Computing Center, San Antonio, Texas, USAEndocrinology DivisionSanta Casa Hospital, São Paulo, São Paulo, BrazilLaboratory of Epidemiology and Population SciencesNational Institute on Aging, Bethesda, Maryland, USA
| | - Monize Lazar
- Endocrine Genetics Unit (Laboratório de Investigação Médica/LIM-25) of Hospital das ClínicasUniversity of São Paulo School of Medicine, São Paulo, São Paulo 05403-010, BrazilNursing SchoolSchool of Public HealthHuman Genome Research CenterUniversity of São Paulo, São Paulo, São Paulo, BrazilDivision of GeneticsGenetic Bases of Thyroid Tumors Laboratory, Department of Morphology and GeneticsDivision of EndocrinologyLaboratory of Molecular and Translational Endocrinology, Department of Medicine, Federal University of São Paulo, São Paulo, São Paulo, BrazilBrazilian National Laboratory of BiosciencesCampinas, São Paulo, BrazilCentro Integral Oncológico Clara CampalHospital Universitário Sanchinarro, Madrid, SpainDepartment of GeneticsTexas Biomedical Research Institute, AT&T Genomic Computing Center, San Antonio, Texas, USAEndocrinology DivisionSanta Casa Hospital, São Paulo, São Paulo, BrazilLaboratory of Epidemiology and Population SciencesNational Institute on Aging, Bethesda, Maryland, USA
| | - Osorio Meirelles
- Endocrine Genetics Unit (Laboratório de Investigação Médica/LIM-25) of Hospital das ClínicasUniversity of São Paulo School of Medicine, São Paulo, São Paulo 05403-010, BrazilNursing SchoolSchool of Public HealthHuman Genome Research CenterUniversity of São Paulo, São Paulo, São Paulo, BrazilDivision of GeneticsGenetic Bases of Thyroid Tumors Laboratory, Department of Morphology and GeneticsDivision of EndocrinologyLaboratory of Molecular and Translational Endocrinology, Department of Medicine, Federal University of São Paulo, São Paulo, São Paulo, BrazilBrazilian National Laboratory of BiosciencesCampinas, São Paulo, BrazilCentro Integral Oncológico Clara CampalHospital Universitário Sanchinarro, Madrid, SpainDepartment of GeneticsTexas Biomedical Research Institute, AT&T Genomic Computing Center, San Antonio, Texas, USAEndocrinology DivisionSanta Casa Hospital, São Paulo, São Paulo, BrazilLaboratory of Epidemiology and Population SciencesNational Institute on Aging, Bethesda, Maryland, USA
| | - Tiago J P Sobreira
- Endocrine Genetics Unit (Laboratório de Investigação Médica/LIM-25) of Hospital das ClínicasUniversity of São Paulo School of Medicine, São Paulo, São Paulo 05403-010, BrazilNursing SchoolSchool of Public HealthHuman Genome Research CenterUniversity of São Paulo, São Paulo, São Paulo, BrazilDivision of GeneticsGenetic Bases of Thyroid Tumors Laboratory, Department of Morphology and GeneticsDivision of EndocrinologyLaboratory of Molecular and Translational Endocrinology, Department of Medicine, Federal University of São Paulo, São Paulo, São Paulo, BrazilBrazilian National Laboratory of BiosciencesCampinas, São Paulo, BrazilCentro Integral Oncológico Clara CampalHospital Universitário Sanchinarro, Madrid, SpainDepartment of GeneticsTexas Biomedical Research Institute, AT&T Genomic Computing Center, San Antonio, Texas, USAEndocrinology DivisionSanta Casa Hospital, São Paulo, São Paulo, BrazilLaboratory of Epidemiology and Population SciencesNational Institute on Aging, Bethesda, Maryland, USA
| | - Maria Lucia Lebrao
- Endocrine Genetics Unit (Laboratório de Investigação Médica/LIM-25) of Hospital das ClínicasUniversity of São Paulo School of Medicine, São Paulo, São Paulo 05403-010, BrazilNursing SchoolSchool of Public HealthHuman Genome Research CenterUniversity of São Paulo, São Paulo, São Paulo, BrazilDivision of GeneticsGenetic Bases of Thyroid Tumors Laboratory, Department of Morphology and GeneticsDivision of EndocrinologyLaboratory of Molecular and Translational Endocrinology, Department of Medicine, Federal University of São Paulo, São Paulo, São Paulo, BrazilBrazilian National Laboratory of BiosciencesCampinas, São Paulo, BrazilCentro Integral Oncológico Clara CampalHospital Universitário Sanchinarro, Madrid, SpainDepartment of GeneticsTexas Biomedical Research Institute, AT&T Genomic Computing Center, San Antonio, Texas, USAEndocrinology DivisionSanta Casa Hospital, São Paulo, São Paulo, BrazilLaboratory of Epidemiology and Population SciencesNational Institute on Aging, Bethesda, Maryland, USA
| | - Yeda A O Duarte
- Endocrine Genetics Unit (Laboratório de Investigação Médica/LIM-25) of Hospital das ClínicasUniversity of São Paulo School of Medicine, São Paulo, São Paulo 05403-010, BrazilNursing SchoolSchool of Public HealthHuman Genome Research CenterUniversity of São Paulo, São Paulo, São Paulo, BrazilDivision of GeneticsGenetic Bases of Thyroid Tumors Laboratory, Department of Morphology and GeneticsDivision of EndocrinologyLaboratory of Molecular and Translational Endocrinology, Department of Medicine, Federal University of São Paulo, São Paulo, São Paulo, BrazilBrazilian National Laboratory of BiosciencesCampinas, São Paulo, BrazilCentro Integral Oncológico Clara CampalHospital Universitário Sanchinarro, Madrid, SpainDepartment of GeneticsTexas Biomedical Research Institute, AT&T Genomic Computing Center, San Antonio, Texas, USAEndocrinology DivisionSanta Casa Hospital, São Paulo, São Paulo, BrazilLaboratory of Epidemiology and Population SciencesNational Institute on Aging, Bethesda, Maryland, USA
| | - John Blangero
- Endocrine Genetics Unit (Laboratório de Investigação Médica/LIM-25) of Hospital das ClínicasUniversity of São Paulo School of Medicine, São Paulo, São Paulo 05403-010, BrazilNursing SchoolSchool of Public HealthHuman Genome Research CenterUniversity of São Paulo, São Paulo, São Paulo, BrazilDivision of GeneticsGenetic Bases of Thyroid Tumors Laboratory, Department of Morphology and GeneticsDivision of EndocrinologyLaboratory of Molecular and Translational Endocrinology, Department of Medicine, Federal University of São Paulo, São Paulo, São Paulo, BrazilBrazilian National Laboratory of BiosciencesCampinas, São Paulo, BrazilCentro Integral Oncológico Clara CampalHospital Universitário Sanchinarro, Madrid, SpainDepartment of GeneticsTexas Biomedical Research Institute, AT&T Genomic Computing Center, San Antonio, Texas, USAEndocrinology DivisionSanta Casa Hospital, São Paulo, São Paulo, BrazilLaboratory of Epidemiology and Population SciencesNational Institute on Aging, Bethesda, Maryland, USA
| | - Mayana Zatz
- Endocrine Genetics Unit (Laboratório de Investigação Médica/LIM-25) of Hospital das ClínicasUniversity of São Paulo School of Medicine, São Paulo, São Paulo 05403-010, BrazilNursing SchoolSchool of Public HealthHuman Genome Research CenterUniversity of São Paulo, São Paulo, São Paulo, BrazilDivision of GeneticsGenetic Bases of Thyroid Tumors Laboratory, Department of Morphology and GeneticsDivision of EndocrinologyLaboratory of Molecular and Translational Endocrinology, Department of Medicine, Federal University of São Paulo, São Paulo, São Paulo, BrazilBrazilian National Laboratory of BiosciencesCampinas, São Paulo, BrazilCentro Integral Oncológico Clara CampalHospital Universitário Sanchinarro, Madrid, SpainDepartment of GeneticsTexas Biomedical Research Institute, AT&T Genomic Computing Center, San Antonio, Texas, USAEndocrinology DivisionSanta Casa Hospital, São Paulo, São Paulo, BrazilLaboratory of Epidemiology and Population SciencesNational Institute on Aging, Bethesda, Maryland, USA
| | - Janete M Cerutti
- Endocrine Genetics Unit (Laboratório de Investigação Médica/LIM-25) of Hospital das ClínicasUniversity of São Paulo School of Medicine, São Paulo, São Paulo 05403-010, BrazilNursing SchoolSchool of Public HealthHuman Genome Research CenterUniversity of São Paulo, São Paulo, São Paulo, BrazilDivision of GeneticsGenetic Bases of Thyroid Tumors Laboratory, Department of Morphology and GeneticsDivision of EndocrinologyLaboratory of Molecular and Translational Endocrinology, Department of Medicine, Federal University of São Paulo, São Paulo, São Paulo, BrazilBrazilian National Laboratory of BiosciencesCampinas, São Paulo, BrazilCentro Integral Oncológico Clara CampalHospital Universitário Sanchinarro, Madrid, SpainDepartment of GeneticsTexas Biomedical Research Institute, AT&T Genomic Computing Center, San Antonio, Texas, USAEndocrinology DivisionSanta Casa Hospital, São Paulo, São Paulo, BrazilLaboratory of Epidemiology and Population SciencesNational Institute on Aging, Bethesda, Maryland, USA
| | - Rui M B Maciel
- Endocrine Genetics Unit (Laboratório de Investigação Médica/LIM-25) of Hospital das ClínicasUniversity of São Paulo School of Medicine, São Paulo, São Paulo 05403-010, BrazilNursing SchoolSchool of Public HealthHuman Genome Research CenterUniversity of São Paulo, São Paulo, São Paulo, BrazilDivision of GeneticsGenetic Bases of Thyroid Tumors Laboratory, Department of Morphology and GeneticsDivision of EndocrinologyLaboratory of Molecular and Translational Endocrinology, Department of Medicine, Federal University of São Paulo, São Paulo, São Paulo, BrazilBrazilian National Laboratory of BiosciencesCampinas, São Paulo, BrazilCentro Integral Oncológico Clara CampalHospital Universitário Sanchinarro, Madrid, SpainDepartment of GeneticsTexas Biomedical Research Institute, AT&T Genomic Computing Center, San Antonio, Texas, USAEndocrinology DivisionSanta Casa Hospital, São Paulo, São Paulo, BrazilLaboratory of Epidemiology and Population SciencesNational Institute on Aging, Bethesda, Maryland, USA
| | - Sergio P A Toledo
- Endocrine Genetics Unit (Laboratório de Investigação Médica/LIM-25) of Hospital das ClínicasUniversity of São Paulo School of Medicine, São Paulo, São Paulo 05403-010, BrazilNursing SchoolSchool of Public HealthHuman Genome Research CenterUniversity of São Paulo, São Paulo, São Paulo, BrazilDivision of GeneticsGenetic Bases of Thyroid Tumors Laboratory, Department of Morphology and GeneticsDivision of EndocrinologyLaboratory of Molecular and Translational Endocrinology, Department of Medicine, Federal University of São Paulo, São Paulo, São Paulo, BrazilBrazilian National Laboratory of BiosciencesCampinas, São Paulo, BrazilCentro Integral Oncológico Clara CampalHospital Universitário Sanchinarro, Madrid, SpainDepartment of GeneticsTexas Biomedical Research Institute, AT&T Genomic Computing Center, San Antonio, Texas, USAEndocrinology DivisionSanta Casa Hospital, São Paulo, São Paulo, BrazilLaboratory of Epidemiology and Population SciencesNational Institute on Aging, Bethesda, Maryland, USA Endocrine Genetics Unit (Laboratório de Investigação Médica/LIM-25) of Hospital das ClínicasUniversity of São Paulo School of Medicine, São Paulo, São Paulo 05403-010, BrazilNursing SchoolSchool of Public HealthHuman Genome Research CenterUniversity of São Paulo, São Paulo, São Paulo, BrazilDivision of GeneticsGenetic Bases of Thyroid Tumors Laboratory, Department of Morphology and GeneticsDivision of EndocrinologyLaboratory of Molecular and Translational Endocrinology, Department of Medicine, Federal University of São Paulo, São Paulo, São Paulo, BrazilBrazilian National Laboratory of BiosciencesCampinas, São Paulo, BrazilCentro Integral Oncológico Clara CampalHospital Universitário Sanchinarro, Madrid, SpainDepartment of GeneticsTexas Biomedical Research Institute, AT&T Genomic Computing Center, San Antonio, Texas, USAEndocrinology DivisionSanta Casa Hospital, São Paulo, São Paulo, BrazilLaboratory of Epidemiology and Population SciencesNat
| |
Collapse
|