Skip to main content
Erschienen in: BMC Medical Genetics 1/2007

Open Access 01.09.2007 | Research

Genetic correlates of longevity and selected age-related phenotypes: a genome-wide association study in the Framingham Study

verfasst von: Kathryn L Lunetta, Ralph B D'Agostino Sr, David Karasik, Emelia J Benjamin, Chao-Yu Guo, Raju Govindaraju, Douglas P Kiel, Margaret Kelly-Hayes, Joseph M Massaro, Michael J Pencina, Sudha Seshadri, Joanne M Murabito

Erschienen in: BMC Medical Genetics | Sonderheft 1/2007

Abstract

Background

Family studies and heritability estimates provide evidence for a genetic contribution to variation in the human life span.

Methods

We conducted a genome wide association study (Affymetrix 100K SNP GeneChip) for longevity-related traits in a community-based sample. We report on 5 longevity and aging traits in up to 1345 Framingham Study participants from 330 families. Multivariable-adjusted residuals were computed using appropriate models (Cox proportional hazards, logistic, or linear regression) and the residuals from these models were used to test for association with qualifying SNPs (70, 987 autosomal SNPs with genotypic call rate ≥80%, minor allele frequency ≥10%, Hardy-Weinberg test p ≥ 0.001).

Results

In family-based association test (FBAT) models, 8 SNPs in two regions approximately 500 kb apart on chromosome 1 (physical positions 73,091,610 and 73, 527,652) were associated with age at death (p-value < 10-5). The two sets of SNPs were in high linkage disequilibrium (minimum r2 = 0.58). The top 30 SNPs for generalized estimating equation (GEE) tests of association with age at death included rs10507486 (p = 0.0001) and rs4943794 (p = 0.0002), SNPs intronic to FOXO1A, a gene implicated in lifespan extension in animal models. FBAT models identified 7 SNPs and GEE models identified 9 SNPs associated with both age at death and morbidity-free survival at age 65 including rs2374983 near PON1. In the analysis of selected candidate genes, SNP associations (FBAT or GEE p-value < 0.01) were identified for age at death in or near the following genes: FOXO1A, GAPDH, KL, LEPR, PON1, PSEN1, SOD2, and WRN. Top ranked SNP associations in the GEE model for age at natural menopause included rs6910534 (p = 0.00003) near FOXO3a and rs3751591 (p = 0.00006) in CYP19A1. Results of all longevity phenotype-genotype associations for all autosomal SNPs are web posted at http://​www.​ncbi.​nlm.​nih.​gov/​projects/​gap/​cgi-bin/​study.​cgi?​id=​phs000007.

Conclusion

Longevity and aging traits are associated with SNPs on the Affymetrix 100K GeneChip. None of the associations achieved genome-wide significance. These data generate hypotheses and serve as a resource for replication as more genes and biologic pathways are proposed as contributing to longevity and healthy aging.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1471-2350-8-S1-S13) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

All authors have made substantial contributions to conception and design or acquisition of phenotypic data. JMM, KL, EJB, CG, DK, DPK, JMM, MJP, RBD contributed to the analysis and interpretation of data. JMM, KL, EJB, DK, DPK, SS have been involved in drafting the manuscript or revising it critically for important intellectual content. All authors read and approved the final manuscript.
Abkürzungen
CVD
cardiovascular disease
FBAT
family-based association test
FHS
Framingham Heart Study
GEE
generalized estimating equations
GWAS
genome-wide association study
LD
linkage disequilibrium
LOD
logarithm of the odds
NCBI
National Center Biotechnology Information
OSS
osseographic scoring system
SNP
single nucleotide polymorphism
TIA
transient ischemic attack.

Background

Genetic factors associated with human longevity and healthy aging remain largely unknown. Heritability estimates of longevity derived from twin registries and large population-based samples suggest a significant but modest genetic contribution to the human lifespan (heritability ~15 to 30%) [14]. However, genetic influences on lifespan may be greater once an individual achieves age 60 years [5]. Moreover, the reported magnitude of the genetic contribution to other important aspects of aging such as healthy physical aging (wellness)[6], physical performance [7, 8], cognitive function [9], and bone aging [10] are much larger. Both exceptional longevity and a healthy aging phenotype have been linked to the same region on chromosome 4 [11, 12], suggesting that although longevity per se and healthy aging are different phenotypes, they may share some common genetic pathways.
A number of potential candidate genes in a variety of biological pathways have been associated with longevity in model organisms. Genes involved in the regulation of DNA repair and genes in the evolutionarily conserved insulin/insulin-like growth factor signaling pathway [13, 14] are emerging as holding great promise in the future elucidation of the underlying physiology controlling lifespan. Many of these genes have human homologs and thus have potential to provide insights into human longevity [1520]. Although numerous candidate genes have been proposed, studies in humans are limited and initial findings often fail replication [21, 22]. More recently genome-wide association studies (GWAS) have become feasible and offer a more comprehensive and untargeted approach to detect genes with modest phenotypic effects that underlie common complex conditions [23].
We had the opportunity to use the Framingham Heart Study (FHS) Affymetrix 100K SNP genotyping resource for a GWAS of longevity and aging-related phenotypes. The FHS offers the unique advantage of a longitudinal family-based community sample with participants who have been well-characterized throughout adulthood with respect to prospectively ascertained risk factors and diseases and continuously followed until death. We report several strategies for 100K SNP associations: 1) a simple low p-value SNP ranking strategy; 2) SNP selection due to associations with more than one related phenotype; and 3) SNP associations within candidate genes and regions previously reported to be associated with longevity in model organisms or humans.

Methods

Study sample

The genotyped study sample is comprised of 1345 Original cohort (n = 258) and Offspring (n = 1087) participants who are members of the 330 largest FHS families. The Overview [24] provides further details of this sample. With respect to aging and longevity traits, 149 deaths occurred at a mean age at death of 83 years (range 46 to 99 years) and 713 participants achieved age 65 years or greater. The Boston University Medical Center Institutional Review Board approved the examination content of Original Cohort and Offspring examinations. All participants provided written informed consent at every examination including consent for genetic studies.

Longevity and aging phenotype definitions and residual creation

Age at death

Both the Original Cohort and the Offspring Cohort remain under continuous surveillance and all deaths that occurred prior to January 1, 2005 were included in this study. Deaths were identified using multiple strategies including routine participant contact for research examinations or health history updates, surveillance at the local hospital, search of obituaries in the local newspaper, and if needed through use of the National Death Index. Death certificates were routinely obtained and all hospital and nursing home records prior to death and autopsy reports (if performed) were requested. In addition, if there was insufficient information to determine a cause of death, the next of kin were interviewed by a senior investigator. All records pertinent to the death were reviewed by an endpoint panel comprised of three senior investigators. The date and cause of death (classified as due to coronary heart disease, stroke, other cardiovascular disease [CVD], cancer, other causes, or unknown cause) was recorded.
Cox proportional hazards models were used to generate martingale residuals using the PHREG procedure in SAS to perform the regression analysis of survival time from age at study entry to age at death. Models were sex-specific and adjusted for 1) birth cohort and 2) birth cohort, education, current smoking status (yes/no), obesity (body mass index ≥30 kg/m2), hypertension (blood pressure ≥140/90 mmHg or on antihypertensive treatment), elevated cholesterol (cholesterol > 239 mg/dL), diabetes (fasting blood sugar ≥126 mg/dL, random blood sugar of ≥200 mg/dL, or use of insulin or oral hypoglycemic agents) and comorbidity defined as CVD and cancer. Birth cohort was defined as a categorical variable for all regression models with the following categories based on year of birth: birth year prior to 1900, 1900 to 1909, 1910 to 1919, 1920 to 1929, 1930 to 1939, 1940 to 1949, and 1950 and later. All covariates were measured at study entry. Residuals from Original Cohort and Offspring participants were pooled.

Morbidity-free survival at age 65 years

Morbidity-free survival was defined as achieving age 65 years free of CVD, dementia, and cancer. CVD events included angina pectoris, coronary insufficiency, myocardial infarction, heart failure, stroke, transient ischemic attack (TIA), intermittent claudication and coronary or CVD death. Suspected CVD events were reviewed by a panel of three investigators who adjudicated events using previously established criteria in place since study inception [25]. A separate panel of study neurologists determined the presence of stroke or TIA and a team of at least one neurologist and one neuropsychologist determined the presence of dementia. Two independent reviewers examined records for all cancers, and the vast majority of cancer cases were microscopically confirmed with pathology reports.
Logistic regression models were used to generate deviance residuals. Models were sex-specific and adjusted for 1) birth cohort and 2) birth cohort, education, current smoking status, obesity, hypertension, elevated cholesterol, and diabetes. Covariates were defined as above for age at death. All covariates were measured at the examination closest to the participant attaining age 65 years using a 5 year window around age 65 years. Residuals from Original Cohort and Offspring participants were pooled.

Age at natural menopause

Natural menopause occurred after a woman had ceased menstruating naturally for one year and the age at natural menopause was the self-reported age at last menstruation. Mean age at natural menopause was similar in Original Cohort and Offspring women and the distribution of naturally menopausal ages in women in the 330 FHS families was similar to that of women in all 1643 FHS families [26, 27]. The mean age at natural menopause in women in the 100K sample was 50.2 years (range 38 to 57 years) in Original Cohort women and 49.1 years (range 29 to 60 years) in Offspring women.
Crude age at natural menopause and standardized residuals from multiple linear regressions in SAS [28] that adjusted age at natural menopause for covariates of interest were used as traits for analysis. Covariates were obtained at all attended examinations prior to the onset of menopause and included mean number of cigarettes smoked per day, mean body mass index, parity (0 versus 1 or more live births), and generation (Original Cohort vs. Offspring).

Walking speed

Walking speed was measured on Original Cohort participants at examination 27 (January 2002 through December 2003, mean age of Original Cohort at exam 27: 86.7 years) and Offspring participants attending an ancillary study to examination 7 (1999 to 2004, mean age at exam: 62.0 years). Trained technicians timed participants walking at their normal pace on a four meter course twice and subsequently asked participants to repeat the course walking at a rapid pace. The mean timed fast walk among Offspring participants in the 100K genotyping sample was 2.44 seconds (standard deviation 0.89). The timed fast walk was used for analysis. Sex-specific linear regression was used to generate residuals adjusted for age and height measured at the time of the walk.

Biologic age by osseographic scoring system

An osseographic scoring system (OSS) was applied to hand radiographs obtained on original cohort (1967 to 1969, mean age 58.7 years) and offspring participants (1992 to 1993, mean age 51.6 years) [10]. Biologic age was then defined as the standardized residual between the OSS predicted age and the actual age. Biologic age defined by this system predicted mortality [10, 29], was very heritable (h2 = 0.57 ± 0.06), and a genome-wide linkage analysis was performed with LOD scores >1.8 present on chromosomes 3q, 11p, 16q, and 21q [10]. Sex- and cohort-specific ranked residuals generated from linear regression of age on log-OSS adjusted for height, body mass index, menopause, and estrogen therapy, were used for analysis.

Genotyping

Affymetrix 100K SNP GeneChip genotyping and the Marshfield STR genotyping performed by the Mammalian Genotyping Service http://​research.​marshfieldclinic​.​org/​genetics are described in the Overview paper [24].

Statistical analysis

The statistical methods for genome-wide linkage and association analyses are described in the Overview [24].

Association

All residual traits described above as well as the additional traits listed in Table 1 were computed using Cox proportional hazards with martingale residuals for survival traits, logistic regression with deviance residuals for dichotomous traits, and linear regression with standard residuals for quantitative traits. The full set of FHS participants with the phenotype were used to create the residuals. The residuals were used to test for association between the genotyped subset of individuals and the SNPs using additive family-based association test (FBAT) and generalized estimating equations (GEE) models as described in the Overview [24]. A total of 70,987 autosomal SNPs met the criteria of genotypic call rate ≥80%, minor allele frequency ≥10%, Hardy-Weinberg test p ≥ 0.001, and ≥10 informative families for FBAT. The number of tests with an FBAT p < 0.001, p < 0.0001, and p < 0.00001 for all phenotypes was similar to what would be expected under the assumptions that the 70,987 tested SNPs were independent and there were no true associations. The GEE tests tended to give an excess of very small p-values over what would be expected under these assumptions.
Table 1
Aging and Longevity Phenotypes for Framingham Heart Study 100K Project
   
Exam cycle(s)
 
Phenotype Subgroup
• Trait (variable name on the website*)
Number of Traits
N (MV**)
Offspring / Original Cohort
 
Adjustment
Survival Traits: Cox regression
Survival
• Age at death (1. deathageX, 2. deathageMV)
2
1345 (1166)
Cohort & Offspring pooled
 
Cox regression
Sex-specific
1. birth cohort
2. multivariable adjusted for birth cohort, education, smoking, obesity (BMI ≥ 30), CVD risk factors, co-morbidity measured at exam 1
Categorical traits: Logistic regression
• Survival past the ALE (1. deathpastALEX, 2. deathpastALEMV)
2
1345 (1166)
Cohort & Offspring pooled
 
Logistic regression
Sex-specific
1. birth cohort
2. multivariable adjusted for birth cohort, education, smoking, obesity (BMI ≥ 30), CVD risk factors, co-morbidity measured at exam 1
Morbidity-free survival (free of CVD, cancer and dementia)
• At age 65 years (1. morbidityfree65X, 2. morbidityfree65MVX)
2
558 (558)
Cohort & Offspring pooled, exams closest to age 65 years
 
Logistic regression
Sex-specific
1. birth cohort
2. multivariable adjusted for birth cohort, education, smoking, obesity, CVD risk factors measured at exam closest to age 65 years (within a 5 year horizon)
Quantitative Traits: Linear regression
Reproductive Aging
• Age at natural menopause (1. menoageX, 2. menoageMVX)
2
438 (378)
Cohort & Offspring pooled, women only
 
Linear regression
1. crude
2. multivariable adjusted for smoking, BMI, parity, generation (measured at exams prior to menopause)
Cognitive function
• MMSE at age 65 years (1. MMSE65X, 2. MMSE65MVX)
• MMSE at the specified Offspring exam (1. MMSE5X, 2. MMSE5MVX, 1. MMSE7X, 2. MMSE7MV, 1. MMSE5to7X, 2. MMSE5to7MVX)
2
593 (462)
Cohort & Offspring pooled, exams at age 65
 
Linear regression
Sex-specific
1. birth cohort
2. multivariable adjusted for birth cohort, education, FSRP measured at exam closest to age 65 years (5 year horizon)
 
6
1038 (913)
Exam 5
Exam 7
Exam 5 & 7
average score
 
Linear regression
Sex-specific
1. birth cohort
2. multivariable adjusted for birth cohort, education, FSRP; covariates measured at the specified exam
Physical Performance
• Hand grip (2. handgrip7x, 2. handgrips727x)
• Walking speed (2. walkingspeed7x, 2. walkingspeed727x)
6
764
Exam 7
Exam 7 and Exam 27
 
Linear regression
Sex-specific‡
1. age
2. multivariable adjusted for age, height, weight at the specified exam
Biologic Age by Osseographic Scoring System (1. deltaOSSr, delta OSSrf, deltaOSSrm)
3
714
Offspring and Cohort pooled
exam 6/7 and exam 22
 
Linear regression
Sex- and cohort-specific ranked residuals§
1. multivariable adjusted for age, height, BMI, menopause, estrogen use
Residuals from these models were used as traits to test for association with SNP genotypes.
* The number preceding the variable name refers to the covariate adjustment in the last column of the table. The website with all results is found at http://​www.​ncbi.​nlm.​nih.​gov/​projects/​gap/​cgi-bin/​study.​cgi?​id=​phs000007; ** MV = N for multivariable trait
‡ cohort- and sex-specific residuals for traits that included both cohort and offspring; §cohort-specific for traits limited to one sex
ALE = average life expectancy, BMI = body mass index, Co-morbidity = cardiovascular disease and cancer, CVD = cardiovascular disease, FSRP = Framingham stroke risk profile, MMSE = mini-mental state exam, Risk factors = hypertension, diabetes, elevated cholesterol

SNP prioritization

We used several strategies to prioritize SNPs associated with longevity and aging traits. First, we used an untargeted approach whereby the top 50 SNP associations ranked according to the strength of the p-value for each trait were examined. Next, we explored the consistency of SNP associations across related sets of traits chosen a priori (trait set one: age at death and morbidity-free survival at age 65 years; trait set two: biologic age and walking speed). Trait set one was chosen based upon linkage data in humans demonstrating that both longevity and a healthy aging trait were linked to the same region on chromosome 4 raising the hypothesis that the two phenotypes may share common genetic pathways [11, 12]. The traits in set two reflect aging with good physical functioning and thus we postulated that biologic age and walking speed may have genetic variants in common. We also investigated SNP associations in candidate genes and regions reported to be associated with longevity identified from established databases including NCBI [14] using the search term "longevity" and the Science of Aging Knowledge Environment genes/intervention database http://​sageke.​sciencemag.​org/​cgi/​genesdb[30] choosing genes potentially related to lifespan in humans.
The SNPs were annotated using the UCSC genome browser tables using the May 2004 assembly http://​genome.​ucsc.​edu/​[31, 32]. All genes within 60 kb of the top ranked SNPs were identified.

Results

The longevity and aging traits available in the FHS 100K SNP resource are listed in Table 1. In this report, we consider only five of the traits listed in Table 1: multivariable-adjusted age at death, morbidity-free survival at age 65 years, age at natural menopause, walking speed, and biologic age by OSS. These traits include a pooled sample of Original Cohort and Offspring participants, with the exception of walking speed, which is reported in Offspring participants only. Details of the sample size and covariate adjustment for each trait are provided in Table 1.
For each of the five phenotypes, Table 2a and 2b provides the top five SNPs ranked in order by lowest p-value for the GEE and FBAT models (all associations can be viewed on the web http://​www.​ncbi.​nlm.​nih.​gov/​projects/​gap/​cgi-bin/​study.​cgi?​id=​phs000007). If multiple SNPs in linkage disequilibrium (LD r2 > 0.80) were included in the top 5, additional SNPs were included until a set of 5 independent associations were listed. Eight SNPs on chromosome 1 were associated with age at death in the FBAT analysis; all with p-value < 10-4 and two with p-value < 10-5. The 8 SNPs consisted of two sets of SNPs (rs10493513, rs10493514, rs6689491, rs6657082, rs1405051) and (rs10493515, rs10493518, rs10493517), clustered in two regions approximately 500 kb apart. There was exceptionally high LD across this 500 kb region: the minimum r2 between pairs of the eight SNPs was 0.58. The nearest genes in this region existing in public databases were >500 kb from any of these SNPs [31, 32].
Table 2
Aging and Longevity Phenotypes† for FHS 100K Project: Results of Association and Linkage Analyses
2a. GEE, Top 5 p-values by Phenotype*
Trait
SNP
Chromosome
Physical location
GEE p-value
FBAT p-value
Gene Region (within 60 kb)
Age at death
 
rs1528753
11
90,523,987
8.1 × 10 -8
0.024
 
 
rs2371208
7
81,982,510
2.6 × 10 -6
0.031
 
 
rs10496799
2
139,261,401
1.4 × 10 -5
0.735
NXPH2
 
rs10489006
4
31,444,987
3.6 × 10 -5
0.078
 
 
rs3757354
6
16,235,386
6.4 × 10 -5
0.316
MYLIP
Morbidity-free survival at age 65
 
rs1412337
1
165,350,299
1.8 × 10 -9
0.505
DPT
 
rs32566
5
5,845,507
1.9 × 10 -9
0.323
 
 
rs10484246
6
9,559,183
8.4 × 10 -8
0.928
 
 
rs4831837
8
12,756,234
4.7 × 10 -7
0.182
 
 
rs2639889
16
59,680,648
9.4 × 10 -7
0.903
 
Age at natural menopause‡
 
rs10496265
2
81,580,466
1.1 × 10 -8
0.001
 
 
rs10496262*
2
81,662,782
3.3 × 10 -7
0.005
 
 
rs958672
2
154,896,075
1.9 × 10 -6
0.087
GALNT13
 
rs291353
1
232,046,939
5.5 × 10 -6
0.035
GNG4
 
rs726336
5
163,911,906
1.1 × 10 -5
0.125
 
Walking speed exam 7
 
rs7137869
12
118,452,366
6.3 × 10 -7
0.009
CCDC60
 
rs7662116
4
154,375,569
1.9 × 10 -5
0.016
 
 
rs7972859*
12
118,452,765
2.5 × 10 -5
0.005
CCDC60
 
rs9318312
13
74,489,506
5.8 × 10 -5
0.266
 
 
rs1994854
4
78,124,824
9.4 × 10 -5
0.280
 
 
rs7718104
5
122,183,258
1.2 × 10 -4
0.011
SNX2
Biologic age by osseographic scoring system
 
rs1463605
12
30,005,150
7.0 × 10 -8
5.3 × 10-4
 
 
rs7176093
15
84,170,434
7.4 × 10 -6
0.005
KLHL25
 
rs3772255
3
157,585,436
8.2 × 10 -6
0.085
KCNAB1
 
rs726846
5
136,099,953
1.1 × 10 -5
0.003
 
 
rs646983
13
29,413,553
1.2 × 10 -5
0.003
 
2b. FBAT, Top 5 p-values by Phenotype*
Trait
SNP
Chromosome
Physical location
GEE p-value
FBAT p-value
Gene Region (within 60 kb)
Age at death
 
rs10493513
1
73,091,610
0.640
1.5 × 10 -6
 
 
rs10493514*
1
73,092,533
0.623
2.8 × 10 -6
 
 
rs6689491*
1
73,064,050
0.205
2.0 × 10 -5
 
 
rs10493515
1
73,527,652
0.225
2.3 × 10 -5
 
 
rs10493518*
1
73,572,652
0.191
3.6 × 10 -5
 
 
rs10493517*
1
73,570,372
0.215
4.2 × 10 -5
 
 
rs6657082*
1
73,065,349
0.224
5.5 × 10 -5
 
 
rs10498263
14
19,285,288
0.310
8.3 × 10 -5
OR4Q3
OR4M1
 
rs1915501
4
28,612,632
0.383
1.1 × 10 -4
 
 
rs1405051*
1
73,060,505
0.176
1.4 × 10 -4
 
 
rs6459623
6
18,634,791
0.604
1.5 × 10 -4
IBRDC2
Morbidity-free survival at age 65
 
rs10509200
10
65,296,567
0.613
7.0 × 10 -5
 
 
rs965036
6
20,099,022
0.550
8.6 × 10 -5
 
 
rs720565
6
136,834,657
0.014
9.8 × 10 -5
MAP7
 
rs1192372
2
84,923,204
0.094
9.9 × 10 -5
 
 
rs10505239
8
115,976,403
0.141
1.3 × 10 -4
 
Age at natural menopause†
 
rs959702
10
2,139,260
0.003
1.4 × 10 -5
 
 
rs7165378
15
69,478,558
0.006
6.4 × 10 -5
 
 
rs997161
10
130,876,127
0.074
8.6 × 10 -5
 
 
rs165284
1
91,235,574
0.006
8.6 × 10 -5
ZNF644
 
rs2280585
3
64,882,324
0.884
1.1 × 10 -4
 
Walking speed exam 7
 
rs4471448
11
86,760,972
0.175
3.8 × 10 -6
TMEM135
 
rs336963
5
83,005,460
0.570
2.6 × 10 -5
HAPLN1
 
rs7862683
9
18,257,947
0.001
4.9 × 10 -5
 
 
rs9317757
13
68,458,430
0.252
7.8 × 10 -5
 
 
rs10501636*
11
86,789,967
0.355
1.1 × 10 -4
 
 
rs2340392
3
80,440,990
0.002
1.7 × 10 -4
 
Biologic age by osseographic scoring system
 
rs1380703
2
57,852,938
0.008
1.1 × 10 -5
 
 
rs324702
4
77,094,969
0.390
3.3 × 10 -5
PPEF2
 
rs324735
4
77,062,193
0.096
7.6 × 10 -5
 
 
rs1106184
2
10,914,125
0.006
8.8 × 10 -5
PDIA6
 
rs604578
18
30,923,438
0.004
9.5 × 10 -5
MAPRE2
2c Linkage Peaks with LOD scores ≥ 2.0
Trait
SNP closest to linkage peak
Chromosome
Physical location
1.5 – LOD support interval start
1.5 – LOD support interval end
LOD score
Age at natural menopause
 
rs1371217
4
182,890,808
178,671,796
186,905,362
2.08
 
rs10509024
10
56,567,832
36,084,470
70,573,011
2.39
 
rs4793513
17
66,429,892
60,635,492
69,512,021
2.48
Walking speed, exam 7
 
rs2769261
1
113,278,949
107,080,550
144,332,709
2.30
 
rs921055
2
233,522,842
229,328,008
242,141,304
2.13
 
rs2602044
3
109,427,883
102,255,525
111,604,019
3.38
 
rs8011773
14
96,927,485
96,342,135
100,389,787
2.69
 
rs1362626
16
4,489,227
205,160
10,344,522
2.05
Biologic age by osseographic scoring system
 
rs353810
9
86,258,745
81,441,976
92,220,168
3.26
 
rs1203981
16
205,160
205,160
7,431,239
2.49
 
rs2248383
21
35,151,811
27,412,716
40,940,879
2.22
SNP criteria: Autosomal SNPs with genotypic call rate ≥ 80%, minor allele frequency ≥ 10%, Hardy-Weinberg test p > 0.001, and ≥10 informative families for FBAT
* For each phenotype SNPs are ranked by p-value. A SNP in LD (r2 > 0.8) with a higher ranked SNP, is identified with an asterisk. All SNPs for a phenotype are listed until 5 independent SNPs are identified. Thus, for some phenotypes more than 5 SNPs are listed. For the age at death trait, the FBAT analysis identified two areas on chromosome 1 in LD, with r2 = .5–.6 between the two regions and r2 of nearly 1.0 within the region.
† Multivariable-adjusted trait results are presented
‡Trait had <500 participants in the sample.
¶Results limited to traits presented
There were several additional associations not listed in Table 2a and 2b that were of interest. For age at death in the GEE analysis, SNP associations ranked numbers 9 and 13 were rs10507486 (p-value 0.000128) and rs4943794 (p-value 0.000277), both are intronic FOXO1A SNPs. For age at natural menopause, top ranked SNP associations in the GEE model included number 11, rs6910534 (p = 0.00003) near FOXO3a and number 18, rs3751591 (p = 0.00006) in CYP19A1.
Table 2c presents the LOD scores ≥2.0 and the corresponding 1.5-LOD support interval from genome-wide linkage for the three quantitative aging traits. None of the regions overlapped with SNPs associated with these aging traits in the FBAT and GEE analyses. Of note for biologic age by OSS the linkage peak on chromosome 21 confirmed a prior Framingham Study report using a genome-wide scan with 401 microsatellite markers [10].
Table 3 provides all SNP associations with a GEE or FBAT p < 0.01 for both traits within the two pairs of related traits. For age at death and morbidity-free survival at age 65 years, FBAT models identified 7 SNPs and GEE models identified 9 SNPs associated with both traits including rs2374983 near PON1 (Tables 3a and 3b). For biologic age by OSS and walking speed, 13 SNPs in FBAT models and 6 SNPs in GEE models were associated with both traits (Tables 3c and 3d).
Table 3
All Significant SNP Associations (GEE or FBAT p-value < 0.01) for at least Two Traits
3a. FBAT: Age at Death and Morbidity-Free Survival at 65 years
Trait 1
Trait 2
SNP
Chr
Physical Position
Gene
Trait 1 GEE p-value
Trait 1 FBAT p-value
Trait 2 GEE p-value
Trait 2 FBAT p-value
Age at Death
Morbidity-free at 65
rs6682403
1
234,743,324
 
0.849
0.004
0.106
0.004
  
rs10488907
4
113,669,709
ALPK1
0.452
0.008
0.336
0.009
  
rs17190837
9
13,391,548
 
0.010
0.009
0.097
0.004
  
rs4752977
11
47,257,005
MADD
0.736
0.008
0.002
0.009
  
rs10506274
12
80,103,932
 
0.531
0.001
0.989
0.001
  
rs2831154
21
28,059,331
 
0.323
0.008
0.358
0.006
  
rs243725*
21
28,060,803
 
0.264
0.007
0.359
0.008
*r2 > 0.80 with the preceding SNP
3b. GEE: Age at Death and Morbidity-Free Survival at 65 years
Trait 1
Trait 2
SNP
Chr
Physical Position
Gene
Trait 1 GEE p-value
Trait 1 FBAT p-value
Trait 2 GEE p-value
Trait 2 FBAT p-value
Age at Death
Morbidity-free at 65
rs9308261
1
113,603,160
MAGI3
0.009
0.156
0.002
0.900
  
rs10490518
2
31,223,004
GALNT14
0.009
0.373
0.010
0.948
  
rs2374983
7
94,516,375
PPP1R9A/PON1
0.006
0.980
0.007
0.727
  
rs655883
11
98,994,584
CNTN5
0.006
0.390
0.001
0.293
  
rs1368850
11
130,433,518
 
0.004
0.387
0.005
0.292
  
rs4943116
13
32,995,650
STARD13
0.006
0.205
0.008
0.049
  
rs2254191
13
45,344,403
 
0.007
0.425
0.004
0.116
  
rs1620210
13
45,759,488
C13orf18
0.001
0.161
0.004
0.068
  
rs2823322
21
15,814,903
 
0.0004
0.045
0.006
0.029
3c. FBAT: Biologic Age and Walking Speed
Trait 1
Trait 2
SNP
Chr
Physical Position
Gene
Trait 1 GEE p-value
Trait 1 FBAT p-value
Trait 2 GEE p-value
Trait 2 FBAT p-value
Biologic age
Walking speed
rs873348
4
178,246,509
 
0.135
0.004
0.114
0.006
Biologic age
Walking speed
rs10520361*
4
178,247,037
 
0.074
0.006
0.054
0.005
Biologic age
Walking speed
rs31564
5
135,258,152
IL9
0.015
0.001
0.008
0.002
Biologic age
Walking speed
rs1862345
5
148,018,498
HTR4
0.172
0.0002
0.399
0.008
Biologic age
Walking speed
rs7844834
8
11,323,556
C8orf12|C8orf13
0.017
0.004
0.011
0.004
Biologic age
Walking speed
rs952658
12
20,756,568
SLCO1C1
0.024
0.008
0.935
0.006
Biologic age
Walking speed
rs6487366
12
23,994,617
SOX5
0.017
0.003
0.959
0.004
Biologic age
Walking speed
rs7135493
12
28,134,847
 
0.020
0.006
0.033
0.004
Biologic age
Walking speed
rs10492036
12
124,728,934
 
0.308
0.005
0.169
0.009
Biologic age
Walking speed
rs1978945
13
105,641,257
 
0.093
0.009
0.031
0.006
Biologic age
Walking speed
rs2165723*
13
105,641,610
 
0.046
0.010
0.027
0.003
Biologic age
Walking speed
rs10492651*
13
105,641,634
 
0.083
0.009
0.023
0.001
Biologic age
Walking speed
rs9301112*
13
105,642,018
 
0.097
0.004
0.053
0.003
* r2 > 0.8 with the preceding SNP (calculated if the distance is <250,000 base pairs)
3d. GEE: Biologic Age and Walking Speed
Trait 1
Trait 2
SNP
Chr
Physical Position
Gene
Trait 1 GEE p-value
Trait 1 FBAT p-value
Trait 2 GEE p-value
Trait 2 FBAT p-value
Biologic Age
Walking speed
rs1474827
6
134,886,011
 
0.007
0.746
0.000
0.233
Biologic Age
Walking speed
rs10231641
7
119,166,342
 
0.004
0.077
0.009
0.278
Biologic Age
Walking speed
rs310575
8
51,603,257
SNTG1
0.008
0.209
0.003
0.007
Biologic Age
Walking speed
rs10520603
15
84,170,955
 
0.009
0.008
0.003
0.050
Biologic Age
Walking speed
rs7166323*
15
84,171,745
 
0.009
0.012
0.003
0.065
Biologic Age
Walking speed
rs2215921
16
9,604,834
 
0.007
0.049
0.001
0.316
* r2 > 0.8 with the preceding SNP (calculated if the distance is <250,000 base pairs)
We identified from the literature 79 potential candidate genes and regions associated with longevity (see Additional file 1 for listing). Of these, 12 genes had no SNPs and 67 genes had 1 to 45 SNPs within 60 kb of the gene on the 100K Affymetrix GeneChip. There were 2036 SNPs in the LGV1 region on chromosome 4 previously linked to exceptional longevity [11]. Table 4 shows the candidate genes with SNPs associated with an FBAT or GEE p-value < 0.01 for age at death including: FOXO1a, GAPDH, KL, LEPR, PON1, PSEN1, SOD2, and WRN and for morbidity-free survival at 65 years including:GHR, LEPR, MORF4L1, PON1, PTH, and WRN. Biologic age by OSS shared 2 SNPs in common with age at death: rs4943794 intronic to FOXO1a and rs911847 near SOD2.
Table 4
All Significant SNP Associations with Selected Longevity Candidate Genes* (FBAT or GEE p-value < 0.01)
Trait
Gene
SNP
Chr
Physical Position
FBAT p-value
GEE p-value
SNP function
SNP position relative
to gene (up to 60 kb)
Age at death
FOXO1a
rs4943794
13
40,071,408
0.068
0.00028
Intron
in
  
rs10507486
13
40,084,501
0.043
0.00013
Intron
in
 
GAPDH†
rs4764600
12
6,472,241
0.833
0.005
Locus/intron
near
 
KL
rs683907
13
32,522,175
0.009
0.507
Intron
in
  
rs687045
13
32,522,889
0.007
0.712
Intron
in
 
LEPR
rs1475398
1
65,695,278
0.069
0.005
Untranslated
in
  
rs1343981
1
65,757,349
0.031
0.006
Intron
in
  
rs10493379
1
65,757,948
0.015
0.004
Intron
in
  
rs2154380
1
65,769,462
0.004
0.003
Intron
in
  
rs6669117
1
65,773,093
0.050
0.007
Intron
in
 
PON1
rs2374983
7
94,516,375
0.980
0.006
Intron
near
 
PSEN1
rs362356
14
72,708,382
0.005
0.130
Intron
in
 
SOD2
rs911847
6
160,039,379
0.358
0.005
Unknown
near
 
WRN‡
rs2543600
8
30,969,282
0.182
4.2 × 10 -6
Unknown
near
Morbidity-free survival at age 65
GHR
rs719756
5
42,761,386
0.003
0.676
Unknown
near
 
LEPR
rs1171278
1
65,700,167
0.042
0.003
Untranslated
in
  
rs3790426
1
65,755,040
0.460
0.002
Intron
in
 
MORF4L1
rs1383636
15
76,893,275
0.458
0.007
Unknown
near
 
PON1
rs2374983
7
94,516,375
0.727
0.007
Intron
near
  
rs854523
7
94,542,884
0.850
0.007
Intron
in
 
PTH
rs10500784
11
13,530,401
0.010
0.990
Unknown
near
 
WRN‡
rs2725369
8
30,970,566
0.113
0.003
Unknown
near
Biologic Age by OSS
FOXO1a
rs1923249
13
40,041,881
0.006
0.004
Intron
in
  
rs4943794
13
40,071,408
0.009
0.016
Intron
in
 
HSPA9
rs256014
5
137,930,983
0.101
0.005
Intron
in
 
LASS6
rs1002666
2
169,303,525
0.001
0.008
Intron
in
 
SOD2
rs911847
6
160,039,379
0.024
0.009
Unknown
near
 
TLR4
rs1927914
9
117,544,279
0.007
0.401
Locus
near
Walking speed
ESR1
rs9322361
6
152,551,257
0.124
0.0089
Intron
in
 
LASS6
rs6433083
2
169,324,821
0.232
0.006
Intron
in
 
NR3C1
rs2918418
5
142,703,566
0.005
0.081
Intron
in
  
rs10515522
5
142,738,587
0.004
0.084
Intron
in
 
SOD1
rs2833485
21
32,000,796
0.008
0.507
Locus/intron
in
 
TERF2
rs728546
16
68,013,029
0.0045
0.533
Unknown
near
 
FASLG
rs6700734
1
169,362,468
0.003
0.029
Intron
in
*79 genes identified from NCBI, SAGE ke, and GenAge databases; 12 genes with no SNPs on 100K chip; 67 genes with 1–45 SNPs on 100K chip; LGV1 2036 SNPs on 100K chip, results for this region available on the web
†The most strongly associated SNP near GAPDH is actually closer to MRPL51
‡The most strongly associated SNP near WRN is actually closer to PURG

Discussion

To our knowledge, this is the first dense GWAS of longevity and aging traits in a community-based sample of adults from two generations of the same families. Over 1300 men and women have detailed longevity and aging-related phenotypes and 100K SNP genotyping results available on the web. This resource has the potential to detect novel susceptibility genes for human longevity and aging and to examine the relevance of promising candidate gene associations reported in animal models to human aging. We describe several strategies to prioritize SNP associations in this unique resource to enhance the discovery of various genes and pathways that contribute to the control of human longevity. Furthermore, FHS investigators are part of the NIA sponsored Longevity Consortium http://​www.​longevityconsort​ium.​org which offers the opportunity of collaboration with other investigators to replicate important findings in additional cohorts.
In our untargeted approach of ranking SNP associations by the strength of the p-value, 2 intronic FOXO1a SNPs were associated with age at death. One of these SNPs (rs4943794) also was associated with biologic age by OSS in our a priori evaluation of select candidate genes. FOXO (forkhead box group O) transcription factors are targets of insulin-like signaling and are involved in a diverse set of physiological functions including DNA repair and resistance to oxidative stress [33, 34]. Further, FOXO plays a role in lifespan extension in C. elegans and Drosophila [35]. Studies of this gene in humans are limited; two case-control studies have not identified an association between FOXO1a and longevity [36, 37]. However, the prospective population-based Leiden 85-plus Study found that FOXO1a was associated with increased mortality attributable to diabetes related deaths in participants aged 85 years and older [38]. The Leiden 85-plus Study also reported that genetic variation causing a reduction in insulin/IGF-1 signaling resulted in improved old age survival among women [20]. However, that report examined other genes in the insulin/insulin-like signaling pathway and did not specifically examine FOXO1a. Finally, the untargeted approach to SNP selection also identified a SNP near FOXO3a associated with age at natural menopause. This gene has been implicated in oocyte death, depletion of functioning ovarian follicles, and infertility in mice [39, 40] and thus represents a plausible candidate gene for menopause. Most positive common gene variant-disease association studies have failed replication [41] including reports on exceptional longevity. Haplotype-based fine mapping of the region on chromosome 4 linked to human longevity initially suggested the MTP gene, a gene important in lipoprotein synthesis, was associated with longevity [21]. However, this association failed replication in a French cohort of long-lived individuals and subsequent case-control studies of nonagenarians [22, 42]. Beekman, et al [43] found neither linkage to chromosome 4 nor association with the MTP gene and longevity among nonagenarians in the Leiden Longevity Study. Meta-analyses implicated admixture of the control sample in the original report as an explanation for the presumed false-positive association. Thus, our findings are hypothesis generating and their importance can not be determined without evidence of consistent replication in other populations.
We examined pleiotropic effects by identifying SNP associations across two pairs of related traits. One SNP near PON1 emerged as associated with both age at death and morbidity-free survival. Surprisingly, there were relatively few SNPs associated with both traits; prior work had suggested that longevity per se and healthy aging may share common genetic pathways [11, 12]. However, morbidity-free survival was measured at age 65 years, it is possible that as our participants age morbidity-free survival defined at age 75 or 85 years will share additional SNP associations with our longevity trait, age at death. A SNP near SOX5, a gene potentially related to musculoskeletal function was associated with both biologic age by OSS and walking speed.
Our strategy of selecting SNPs in candidate genes and regions previously reported to be associated with longevity yielded interesting findings. For age at death, we identified SNPs in or near several genes including KL, LEPR, PON1, SOD2, and WRN. Defects in the WRN gene are the cause of Werner Syndrome, an autosomal recessive disorder characterized by premature aging. A longitudinal study of ageing Danish twins recently reported a possible association between a successful aging trait and 3 SNPs in the WRN gene [44]. We were unable to determine if our SNP (rs2725369) was in LD with the SNPs in the prior report because the SNPs were not included in HapMap. Mutations in the KL (Klotho) gene in the mouse lead to a syndrome resembling human aging [4547]. There has been one report linking a functional variant of the KL gene to human longevity [15]. Thus, results from this GWAS may direct resources to the most relevant candidate genes and pathways for further investigation in humans.
Several important limitations merit comment. First, we acknowledge that there may be a survival bias as participants in this sample had to survive to provide DNA (first systematic DNA collection began 1995) and hence are likely healthier than the full FHS sample. To ameliorate this issue, we adjusted for covariates using the full Framingham sample, and used the residual traits for the subset of individuals genotyped using the 100K Affymetrix GeneChip to test for association with the SNPs using linear regression models. Residual traits from Cox and logistic models typically are not ideally distributed for linear regression models, but our adjustment method using the full sample precludes the testing of SNP associations with age at death and morbidity-free survival using Cox and logistic models. Second, the 100K Affymetrix GeneChip provides limited coverage of the genome; many of our a priori candidate genes did not have any SNP coverage on the chip. For example, several genes that have been studied in model organisms or even in humans such as ACE, Lamin A, SIRT2 and SIRT3, had no SNPs within 60 kb of the gene on the 100K Affymetrix GeneChip. However, genotyping is near-complete for the NHLBI funded 550 K genome-wide scan on all FHS participants. This will enable deeper exploration of our initial 100K SNP associations in a larger sample with denser coverage of the genome. Third, in this analysis we did not examine epistasis or gene-environment interactions which may modify the associations in this study. Importantly, this study is hypothesis generating. Our findings need to be replicated in other samples.

Conclusion

In summary, the untargeted genome-wide approach to detect genetic associations with longevity and aging traits provides an opportunity to identify novel biologic pathways related to lifespan control. GWAS also have the potential to direct investigators of human aging to the most promising candidate gene associations and biologic pathways reported to regulate lifespan in animal models. Enhancing our understanding of the mechanisms responsible for aging may in turn identify directions for health promotion and disease prevention efforts in middle-aged and older adults so that older persons can enjoy more time in good health. These data generate hypotheses regarding novel biologic pathways contributing to longevity and healthy aging and serve as a resource for replication of findings from other population-based samples.

Acknowledgements

NHLBI's Framingham Heart Study is supported by contract number N01-HC-25195. This work was also supported in part by R01 AR/AG 41398 and AG028321.
FHS 100K analyses were conducted using the Boston University.
Linux Cluster for Genetic Analysis (LinGA) funded by the NIH NCRR (National Center for Research Resources) Shared Instrumentation grant 1S10RR163736-01A1 http://​www.​bu.​edu/​dbin/​sph/​departments/​biostatistics/​linga_​publications.​php.
We thank the FHS participants for their ongoing participation and dedication to the study making this work possible.
This article has been published as part of BMC Medical Genetics Volume 8 Supplement 1, 2007: The Framingham Heart Study 100,000 single nucleotide polymorphisms resource. The full contents of the supplement are available online at http://​www.​biomedcentral.​com/​1471-2350/​8?​issue=​S1.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

All authors have made substantial contributions to conception and design or acquisition of phenotypic data. JMM, KL, EJB, CG, DK, DPK, JMM, MJP, RBD contributed to the analysis and interpretation of data. JMM, KL, EJB, DK, DPK, SS have been involved in drafting the manuscript or revising it critically for important intellectual content. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Herskind AM, McGue M, Holm NV, Sorensen TI, Harvald B, Vaupel JW: The heritability of human longevity: a population-based study of 2872 Danish twin pairs born 1870–1900. HumGenet. 1996, 97 (3): 319- Herskind AM, McGue M, Holm NV, Sorensen TI, Harvald B, Vaupel JW: The heritability of human longevity: a population-based study of 2872 Danish twin pairs born 1870–1900. HumGenet. 1996, 97 (3): 319-
2.
Zurück zum Zitat Iachine IA, Holm NV, Harris JR, Begun AZ, Iachina MK, Laitinen M, Kaprio J, Yashin AI: How heritable is individual susceptibility to death? The results of an analysis of survival data on Danish, Swedish and Finnish twins. TwinRes. 1998, 1 (4): 196-10.1375/136905298320566168. Iachine IA, Holm NV, Harris JR, Begun AZ, Iachina MK, Laitinen M, Kaprio J, Yashin AI: How heritable is individual susceptibility to death? The results of an analysis of survival data on Danish, Swedish and Finnish twins. TwinRes. 1998, 1 (4): 196-10.1375/136905298320566168.
3.
Zurück zum Zitat Ljungquist B, Berg S, Lanke J, McClearn GE, Pedersen NL: The effect of genetic factors for longevity: a comparison of identical and fraternal twins in the Swedish Twin Registry. JGerontolA BiolSciMedSci. 1998, 53 (6): M441- Ljungquist B, Berg S, Lanke J, McClearn GE, Pedersen NL: The effect of genetic factors for longevity: a comparison of identical and fraternal twins in the Swedish Twin Registry. JGerontolA BiolSciMedSci. 1998, 53 (6): M441-
4.
Zurück zum Zitat McGue M, Vaupel JW, Holm N, Harvald B: Longevity is moderately heritable in a sample of Danish twins born 1870–1880. JGerontol. 1993, 48 (6): B237-CrossRef McGue M, Vaupel JW, Holm N, Harvald B: Longevity is moderately heritable in a sample of Danish twins born 1870–1880. JGerontol. 1993, 48 (6): B237-CrossRef
5.
Zurück zum Zitat Hjelmborg JV, Iachine I, Skytthe A, Vaupel JW, McGue M, Koskenvuo M, Kaprio J, Pedersen NL, Christensen K: Genetic influence on human lifespan and longevity. HumGenet. 2006, 119 (3): 312-10.1007/s00439-006-0144-y. Hjelmborg JV, Iachine I, Skytthe A, Vaupel JW, McGue M, Koskenvuo M, Kaprio J, Pedersen NL, Christensen K: Genetic influence on human lifespan and longevity. HumGenet. 2006, 119 (3): 312-10.1007/s00439-006-0144-y.
6.
Zurück zum Zitat Reed T, Dick DM: Heritability and validity of healthy physical aging (wellness) in elderly male twins. TwinRes. 2003, 6 (3): 227-10.1375/136905203765693889. Reed T, Dick DM: Heritability and validity of healthy physical aging (wellness) in elderly male twins. TwinRes. 2003, 6 (3): 227-10.1375/136905203765693889.
7.
Zurück zum Zitat Carmelli D, Kelly-Hayes M, Wolf PA, Swan GE, Jack LM, Reed T, Guralnik JM: The contribution of genetic influences to measures of lower-extremity function in older male twins. JGerontolA BiolSciMedSci. 2000, 55 (1): B49- Carmelli D, Kelly-Hayes M, Wolf PA, Swan GE, Jack LM, Reed T, Guralnik JM: The contribution of genetic influences to measures of lower-extremity function in older male twins. JGerontolA BiolSciMedSci. 2000, 55 (1): B49-
8.
Zurück zum Zitat Reed T, Fabsitz RR, Selby JV, Carmelli D: Genetic influences and grip strength norms in the NHLBI twin study males aged 59–69. AnnHumBiol. 1991, 18 (5): 425- Reed T, Fabsitz RR, Selby JV, Carmelli D: Genetic influences and grip strength norms in the NHLBI twin study males aged 59–69. AnnHumBiol. 1991, 18 (5): 425-
9.
Zurück zum Zitat Swan GE, Carmelli D, Reed T, Harshfield GA, Fabsitz RR, Eslinger PJ: Heritability of cognitive performance in aging twins. The National Heart, Lung, and Blood Institute Twin Study. ArchNeurol. 1990, 47 (3): 259- Swan GE, Carmelli D, Reed T, Harshfield GA, Fabsitz RR, Eslinger PJ: Heritability of cognitive performance in aging twins. The National Heart, Lung, and Blood Institute Twin Study. ArchNeurol. 1990, 47 (3): 259-
10.
Zurück zum Zitat Karasik D, Hannan MT, Cupples LA, Felson DT, Kiel DP: Genetic contribution to biological aging: the Framingham Study. JGerontolA BiolSciMedSci. 2004, 59 (3): 218- Karasik D, Hannan MT, Cupples LA, Felson DT, Kiel DP: Genetic contribution to biological aging: the Framingham Study. JGerontolA BiolSciMedSci. 2004, 59 (3): 218-
11.
Zurück zum Zitat Puca AA, Daly MJ, Brewster SJ, Matise TC, Barrett J, Shea-Drinkwater M, Kang S, Joyce E, Nicoli J, Benson E, Kunkel LM, Perls T: A genome-wide scan for linkage to human exceptional longevity identifies a locus on chromosome 4. ProcNatlAcadSciUSA. 2001, 98 (18): 10505-CrossRef Puca AA, Daly MJ, Brewster SJ, Matise TC, Barrett J, Shea-Drinkwater M, Kang S, Joyce E, Nicoli J, Benson E, Kunkel LM, Perls T: A genome-wide scan for linkage to human exceptional longevity identifies a locus on chromosome 4. ProcNatlAcadSciUSA. 2001, 98 (18): 10505-CrossRef
12.
Zurück zum Zitat Reed T, Dick DM, Uniacke SK, Foroud T, Nichols WC: Genome-wide scan for a healthy aging phenotype provides support for a locus near D4S1564 promoting healthy aging. JGerontolA BiolSciMedSci. 2004, 59 (3): 227- Reed T, Dick DM, Uniacke SK, Foroud T, Nichols WC: Genome-wide scan for a healthy aging phenotype provides support for a locus near D4S1564 promoting healthy aging. JGerontolA BiolSciMedSci. 2004, 59 (3): 227-
13.
Zurück zum Zitat Cheng CL, Gao TQ, Wang Z, Li DD: Role of insulin/insulin-like growth factor 1 signaling pathway in longevity. World JGastroenterol. 2005, 11 (13): 1891-CrossRef Cheng CL, Gao TQ, Wang Z, Li DD: Role of insulin/insulin-like growth factor 1 signaling pathway in longevity. World JGastroenterol. 2005, 11 (13): 1891-CrossRef
14.
Zurück zum Zitat Katic M, Kahn CR: The role of insulin and IGF-1 signaling in longevity. Cell MolLife Sci. 2005, 62 (3): 320-10.1007/s00018-004-4297-y.CrossRef Katic M, Kahn CR: The role of insulin and IGF-1 signaling in longevity. Cell MolLife Sci. 2005, 62 (3): 320-10.1007/s00018-004-4297-y.CrossRef
15.
Zurück zum Zitat Arking DE, Atzmon G, Arking A, Barzilai N, Dietz HC: Association between a functional variant of the KLOTHO gene and high-density lipoprotein cholesterol, blood pressure, stroke, and longevity. CircRes. 2005, 96 (4): 412- Arking DE, Atzmon G, Arking A, Barzilai N, Dietz HC: Association between a functional variant of the KLOTHO gene and high-density lipoprotein cholesterol, blood pressure, stroke, and longevity. CircRes. 2005, 96 (4): 412-
16.
Zurück zum Zitat Bellizzi D, Rose G, Cavalcante P, Covello G, Dato S, De Rango F, Greco V, Maggiolini M, Feraco E, Mari V, Franceschi C, Passarino G, De Benedictis G: A novel VNTR enhancer within the SIRT3 gene, a human homologue of SIR2, is associated with survival at oldest ages. Genomics. 2005, 85 (2): 258-10.1016/j.ygeno.2004.11.003.CrossRefPubMed Bellizzi D, Rose G, Cavalcante P, Covello G, Dato S, De Rango F, Greco V, Maggiolini M, Feraco E, Mari V, Franceschi C, Passarino G, De Benedictis G: A novel VNTR enhancer within the SIRT3 gene, a human homologue of SIR2, is associated with survival at oldest ages. Genomics. 2005, 85 (2): 258-10.1016/j.ygeno.2004.11.003.CrossRefPubMed
17.
Zurück zum Zitat Browner WS, Kahn AJ, Ziv E, Reiner AP, Oshima J, Cawthon RM, Hsueh WC, Cummings SR: The genetics of human longevity. AmJMed. 2004, 117 (11): 851- Browner WS, Kahn AJ, Ziv E, Reiner AP, Oshima J, Cawthon RM, Hsueh WC, Cummings SR: The genetics of human longevity. AmJMed. 2004, 117 (11): 851-
18.
Zurück zum Zitat Hadley EC, Rossi WK: Exceptional survival in human populations: National Institute on Aging perspectives and programs. MechAgeing Dev. 2005, 126 (2): 231- Hadley EC, Rossi WK: Exceptional survival in human populations: National Institute on Aging perspectives and programs. MechAgeing Dev. 2005, 126 (2): 231-
19.
Zurück zum Zitat Rose G, Dato S, Altomare K, Bellizzi D, Garasto S, Greco V, Passarino G, Feraco E, Mari V, Barbi C, Bonafe M, Franceschi C, Tan Q, Boiko S, Yashin AI, De Benedictis G: Variability of the SIRT3 gene, human silent information regulator Sir2 homologue, and survivorship in the elderly. ExpGerontol. 2003, 38 (10): 1065- Rose G, Dato S, Altomare K, Bellizzi D, Garasto S, Greco V, Passarino G, Feraco E, Mari V, Barbi C, Bonafe M, Franceschi C, Tan Q, Boiko S, Yashin AI, De Benedictis G: Variability of the SIRT3 gene, human silent information regulator Sir2 homologue, and survivorship in the elderly. ExpGerontol. 2003, 38 (10): 1065-
20.
Zurück zum Zitat van Heemst D, Beekman M, Mooijaart SP, Heijmans BT, Brandt BW, Zwaan BJ, Slagboom PE, Westendorp RG: Reduced insulin/IGF-1 signalling and human longevity. Aging Cell. 2005, 4 (2): 79-10.1111/j.1474-9728.2005.00148.x.CrossRefPubMed van Heemst D, Beekman M, Mooijaart SP, Heijmans BT, Brandt BW, Zwaan BJ, Slagboom PE, Westendorp RG: Reduced insulin/IGF-1 signalling and human longevity. Aging Cell. 2005, 4 (2): 79-10.1111/j.1474-9728.2005.00148.x.CrossRefPubMed
21.
Zurück zum Zitat Geesaman BJ, Benson E, Brewster SJ, Kunkel LM, Blanche H, Thomas G, Perls TT, Daly MJ, Puca AA: Haplotype-based identification of a microsomal transfer protein marker associated with the human lifespan. ProcNatlAcadSciUSA. 2003, 100 (24): 14115-CrossRef Geesaman BJ, Benson E, Brewster SJ, Kunkel LM, Blanche H, Thomas G, Perls TT, Daly MJ, Puca AA: Haplotype-based identification of a microsomal transfer protein marker associated with the human lifespan. ProcNatlAcadSciUSA. 2003, 100 (24): 14115-CrossRef
22.
Zurück zum Zitat Nebel A, Croucher PJ, Stiegeler R, Nikolaus S, Krawczak M, Schreiber S: No association between microsomal triglyceride transfer protein (MTP) haplotype and longevity in humans. ProcNatlAcadSciUSA. 2005, 102 (22): 7906-CrossRef Nebel A, Croucher PJ, Stiegeler R, Nikolaus S, Krawczak M, Schreiber S: No association between microsomal triglyceride transfer protein (MTP) haplotype and longevity in humans. ProcNatlAcadSciUSA. 2005, 102 (22): 7906-CrossRef
23.
Zurück zum Zitat Hirschhorn JN, Daly MJ: Genome-wide association studies for common diseases and complex traits. NatRevGenet. 2005, 6 (2): 95- Hirschhorn JN, Daly MJ: Genome-wide association studies for common diseases and complex traits. NatRevGenet. 2005, 6 (2): 95-
24.
Zurück zum Zitat Cupples LA, Arruda H, Benjamin EJ, D'Agostino RB, Demissie S, DeStefano AL, Dupuis J, Falls K, Fox CS, Gottlieb DJ, Govindaraju DR, Guo CY, Heard-Costa N, Hwang SJ, Kathiresan S, Kiel DP, Laramie JM, Larson MG, Levy D, Liu CY, Lunetta KL, Mailman MD, Manning AK, Meigs JB, Murabito JM, Newton-Cheh C, O'Connor GT, O'Donnell CJ, Pandey MA, Seshadri S, Vasan RS, Wang ZY, Wilk JB, Wolf PA, Yang Q, Atwood LD: The Framingham Heart Study 100K SNP genome-wide association study resource: Overview of 17 phenotype working group reports. BMC Med Genet. 2007, 8 (Suppl 1): S1-PubMedCentralCrossRefPubMed Cupples LA, Arruda H, Benjamin EJ, D'Agostino RB, Demissie S, DeStefano AL, Dupuis J, Falls K, Fox CS, Gottlieb DJ, Govindaraju DR, Guo CY, Heard-Costa N, Hwang SJ, Kathiresan S, Kiel DP, Laramie JM, Larson MG, Levy D, Liu CY, Lunetta KL, Mailman MD, Manning AK, Meigs JB, Murabito JM, Newton-Cheh C, O'Connor GT, O'Donnell CJ, Pandey MA, Seshadri S, Vasan RS, Wang ZY, Wilk JB, Wolf PA, Yang Q, Atwood LD: The Framingham Heart Study 100K SNP genome-wide association study resource: Overview of 17 phenotype working group reports. BMC Med Genet. 2007, 8 (Suppl 1): S1-PubMedCentralCrossRefPubMed
25.
Zurück zum Zitat Survival following initial cardiovascular disease events: 30 year follow-up. The Framingham Heart Study: An epidemiological investigation of cardiovascular disease. Edited by: Kannel WB, Wolf PA, Garrison RJ. 1988, Bethseda, MD: NHLBI, NIH Survival following initial cardiovascular disease events: 30 year follow-up. The Framingham Heart Study: An epidemiological investigation of cardiovascular disease. Edited by: Kannel WB, Wolf PA, Garrison RJ. 1988, Bethseda, MD: NHLBI, NIH
26.
Zurück zum Zitat Murabito JM, Yang Q, Fox C, Wilson PW, Cupples LA: Heritability of age at natural menopause in the Framingham Heart Study. J Clin Endocrinol Metab. 2005, 90 (6): 3427-3430. 10.1210/jc.2005-0181.CrossRefPubMed Murabito JM, Yang Q, Fox C, Wilson PW, Cupples LA: Heritability of age at natural menopause in the Framingham Heart Study. J Clin Endocrinol Metab. 2005, 90 (6): 3427-3430. 10.1210/jc.2005-0181.CrossRefPubMed
27.
Zurück zum Zitat Murabito JM, Yang Q, Fox CS, Cupples LA: Genome-wide linkage analysis to age at natural menopause in a community-based sample: the Framingham Heart Study. Fertil Steril. 2005, 84 (6): 1674-1679. 10.1016/j.fertnstert.2005.05.046.CrossRefPubMed Murabito JM, Yang Q, Fox CS, Cupples LA: Genome-wide linkage analysis to age at natural menopause in a community-based sample: the Framingham Heart Study. Fertil Steril. 2005, 84 (6): 1674-1679. 10.1016/j.fertnstert.2005.05.046.CrossRefPubMed
28.
Zurück zum Zitat SAS/STAT software, Version 8.2. SAS System for Windows. Copyright 1999–2001. 1999, SAS Institute Inc. Cary, NC, USA SAS/STAT software, Version 8.2. SAS System for Windows. Copyright 1999–2001. 1999, SAS Institute Inc. Cary, NC, USA
29.
Zurück zum Zitat Karasik D, Demissie S, Cupples LA, Kiel DP: Disentangling the genetic determinants of human aging: biological age as an alternative to the use of survival measures. JGerontolA BiolSciMedSci. 2005, 60 (5): 574- Karasik D, Demissie S, Cupples LA, Kiel DP: Disentangling the genetic determinants of human aging: biological age as an alternative to the use of survival measures. JGerontolA BiolSciMedSci. 2005, 60 (5): 574-
31.
Zurück zum Zitat Karolchik D, Baertsch R, Diekhans M, Furey TS, Hinrichs A, Lu YT, Roskin KM, Schwartz M, Sugnet CW, Thomas DJ, Weber RJ, Haussler D, Kent WJ: The UCSC Genome Browser Database. Nucleic Acids Res. 2003, 31 (1): 51-10.1093/nar/gkg129.PubMedCentralCrossRefPubMed Karolchik D, Baertsch R, Diekhans M, Furey TS, Hinrichs A, Lu YT, Roskin KM, Schwartz M, Sugnet CW, Thomas DJ, Weber RJ, Haussler D, Kent WJ: The UCSC Genome Browser Database. Nucleic Acids Res. 2003, 31 (1): 51-10.1093/nar/gkg129.PubMedCentralCrossRefPubMed
32.
Zurück zum Zitat Kent WJ, Sugnet CW, Furey TS, Roskin KM, Pringle TH, Zahler AM, Haussler D: The human genome browser at UCSC. Genome Res. 2002, 12 (6): 996-10.1101/gr.229102. Article published online before print in May 2002.PubMedCentralCrossRefPubMed Kent WJ, Sugnet CW, Furey TS, Roskin KM, Pringle TH, Zahler AM, Haussler D: The human genome browser at UCSC. Genome Res. 2002, 12 (6): 996-10.1101/gr.229102. Article published online before print in May 2002.PubMedCentralCrossRefPubMed
33.
Zurück zum Zitat Barthel A, Schmoll D, Unterman TG: FoxO proteins in insulin action and metabolism. Trends EndocrinolMetab. 2005, 16 (4): 183-10.1016/j.tem.2005.03.010.CrossRef Barthel A, Schmoll D, Unterman TG: FoxO proteins in insulin action and metabolism. Trends EndocrinolMetab. 2005, 16 (4): 183-10.1016/j.tem.2005.03.010.CrossRef
34.
Zurück zum Zitat Lam EW, Francis RE, Petkovic M: FOXO transcription factors: key regulators of cell fate. BiochemSoc Trans. 2006, 34 (Pt 5): 722-CrossRef Lam EW, Francis RE, Petkovic M: FOXO transcription factors: key regulators of cell fate. BiochemSoc Trans. 2006, 34 (Pt 5): 722-CrossRef
35.
Zurück zum Zitat Hwangbo DS, Gershman B, Tu MP, Palmer M, Tatar M: Drosophila dFOXO controls lifespan and regulates insulin signalling in brain and fat body. Nature. 2004, 429 (6991): 562-10.1038/nature02549.CrossRefPubMed Hwangbo DS, Gershman B, Tu MP, Palmer M, Tatar M: Drosophila dFOXO controls lifespan and regulates insulin signalling in brain and fat body. Nature. 2004, 429 (6991): 562-10.1038/nature02549.CrossRefPubMed
36.
Zurück zum Zitat Bonafe M, Barbieri M, Marchegiani F, Olivieri F, Ragno E, Giampieri C, Mugianesi E, Centurelli M, Franceschi C, Paolisso G: Polymorphic variants of insulin-like growth factor I (IGF-I) receptor and phosphoinositide 3-kinase genes affect IGF-I plasma levels and human longevity: cues for an evolutionarily conserved mechanism of life span control. JClinEndocrinolMetab. 2003, 88 (7): 3299- Bonafe M, Barbieri M, Marchegiani F, Olivieri F, Ragno E, Giampieri C, Mugianesi E, Centurelli M, Franceschi C, Paolisso G: Polymorphic variants of insulin-like growth factor I (IGF-I) receptor and phosphoinositide 3-kinase genes affect IGF-I plasma levels and human longevity: cues for an evolutionarily conserved mechanism of life span control. JClinEndocrinolMetab. 2003, 88 (7): 3299-
37.
Zurück zum Zitat Kojima T, Kamei H, Aizu T, Arai Y, Takayama M, Nakazawa S, Ebihara Y, Inagaki H, Masui Y, Gondo Y, Sakaki Y, Hirose N: Association analysis between longevity in the Japanese population and polymorphic variants of genes involved in insulin and insulin-like growth factor 1 signaling pathways. ExpGerontol. 2004, 39 (11–12): 1595- Kojima T, Kamei H, Aizu T, Arai Y, Takayama M, Nakazawa S, Ebihara Y, Inagaki H, Masui Y, Gondo Y, Sakaki Y, Hirose N: Association analysis between longevity in the Japanese population and polymorphic variants of genes involved in insulin and insulin-like growth factor 1 signaling pathways. ExpGerontol. 2004, 39 (11–12): 1595-
38.
Zurück zum Zitat Kuningas M, Magi R, Westendorp RG, Slagboom PE, Remm M, van Heemst D: Haplotypes in the human Foxo1a and Foxo3a genes; impact on disease and mortality at old age. EurJHumGenet. 2007, 15 (3): 294- Kuningas M, Magi R, Westendorp RG, Slagboom PE, Remm M, van Heemst D: Haplotypes in the human Foxo1a and Foxo3a genes; impact on disease and mortality at old age. EurJHumGenet. 2007, 15 (3): 294-
39.
Zurück zum Zitat Brenkman AB, Burgering BM: FoxO3a eggs on fertility and aging. Trends Mol Med. 2003, 9 (11): 464-467. 10.1016/j.molmed.2003.09.003.CrossRefPubMed Brenkman AB, Burgering BM: FoxO3a eggs on fertility and aging. Trends Mol Med. 2003, 9 (11): 464-467. 10.1016/j.molmed.2003.09.003.CrossRefPubMed
40.
Zurück zum Zitat Castrillon DH, Miao L, Kollipara R, Horner JW, DePinho RA: Suppression of ovarian follicle activation in mice by the transcription factor Foxo3a. Science. 2003, 301 (5630): 215-218. 10.1126/science.1086336.CrossRefPubMed Castrillon DH, Miao L, Kollipara R, Horner JW, DePinho RA: Suppression of ovarian follicle activation in mice by the transcription factor Foxo3a. Science. 2003, 301 (5630): 215-218. 10.1126/science.1086336.CrossRefPubMed
41.
Zurück zum Zitat Hirschhorn JN, Lohmueller K, Byrne E, Hirschhorn K: A comprehensive review of genetic association studies. Genet Med. 2002, 4 (2): 45-61.CrossRefPubMed Hirschhorn JN, Lohmueller K, Byrne E, Hirschhorn K: A comprehensive review of genetic association studies. Genet Med. 2002, 4 (2): 45-61.CrossRefPubMed
42.
Zurück zum Zitat Bathum L, Christiansen L, Tan Q, Vaupel J, Jeune B, Christensen K: No evidence for an association between extreme longevity and microsomal transfer protein polymorphisms in a longitudinal study of 1651 nonagenarians. EurJHumGenet. 2005, 13 (10): 1154- Bathum L, Christiansen L, Tan Q, Vaupel J, Jeune B, Christensen K: No evidence for an association between extreme longevity and microsomal transfer protein polymorphisms in a longitudinal study of 1651 nonagenarians. EurJHumGenet. 2005, 13 (10): 1154-
43.
Zurück zum Zitat Beekman M, Blauw GJ, Houwing-Duistermaat JJ, Brandt BW, Westendorp RG, Slagboom PE: Chromosome 4q25, microsomal transfer protein gene, and human longevity: novel data and a meta-analysis of association studies. JGerontolA BiolSciMedSci. 2006, 61 (4): 355- Beekman M, Blauw GJ, Houwing-Duistermaat JJ, Brandt BW, Westendorp RG, Slagboom PE: Chromosome 4q25, microsomal transfer protein gene, and human longevity: novel data and a meta-analysis of association studies. JGerontolA BiolSciMedSci. 2006, 61 (4): 355-
44.
Zurück zum Zitat Sild M, Koca C, Bendixen MH, Frederiksen H, McGue M, Kolvraa S, Christensen K, Nexo B: Possible associations between successful aging and polymorphic markers in the Werner gene region. AnnNYAcadSci. 2006, 1067: 309-CrossRef Sild M, Koca C, Bendixen MH, Frederiksen H, McGue M, Kolvraa S, Christensen K, Nexo B: Possible associations between successful aging and polymorphic markers in the Werner gene region. AnnNYAcadSci. 2006, 1067: 309-CrossRef
45.
Zurück zum Zitat Kuro-o M, Matsumura Y, Aizawa H, Kawaguchi H, Suga T, Utsugi T, Ohyama Y, Kurabayashi M, Kaname T, Kume E, Iwasaki H, Iida A, Shiraki-Iida T, Nishikawa S, Nagai R, Nabeshima YI: Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature. 1997, 390 (6655): 45-10.1038/36285.CrossRefPubMed Kuro-o M, Matsumura Y, Aizawa H, Kawaguchi H, Suga T, Utsugi T, Ohyama Y, Kurabayashi M, Kaname T, Kume E, Iwasaki H, Iida A, Shiraki-Iida T, Nishikawa S, Nagai R, Nabeshima YI: Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature. 1997, 390 (6655): 45-10.1038/36285.CrossRefPubMed
46.
Zurück zum Zitat Kurosu H, Yamamoto M, Clark JD, Pastor JV, Nandi A, Gurnani P, McGuinness OP, Chikuda H, Yamaguchi M, Kawaguchi H, Shimomura I, Takayama Y, Herz J, Kahn CR, Rosenblatt KP, Kuro-o M: Suppression of aging in mice by the hormone Klotho. Science. 2005, 309 (5742): 1829-10.1126/science.1112766.PubMedCentralCrossRefPubMed Kurosu H, Yamamoto M, Clark JD, Pastor JV, Nandi A, Gurnani P, McGuinness OP, Chikuda H, Yamaguchi M, Kawaguchi H, Shimomura I, Takayama Y, Herz J, Kahn CR, Rosenblatt KP, Kuro-o M: Suppression of aging in mice by the hormone Klotho. Science. 2005, 309 (5742): 1829-10.1126/science.1112766.PubMedCentralCrossRefPubMed
47.
Zurück zum Zitat Masuda H, Chikuda H, Suga T, Kawaguchi H, Kuro O: Regulation of multiple ageing-like phenotypes by inducible klotho gene expression in klotho mutant mice. MechAgeing Dev. 2005, 126 (12): 1274- Masuda H, Chikuda H, Suga T, Kawaguchi H, Kuro O: Regulation of multiple ageing-like phenotypes by inducible klotho gene expression in klotho mutant mice. MechAgeing Dev. 2005, 126 (12): 1274-
Metadaten
Titel
Genetic correlates of longevity and selected age-related phenotypes: a genome-wide association study in the Framingham Study
verfasst von
Kathryn L Lunetta
Ralph B D'Agostino Sr
David Karasik
Emelia J Benjamin
Chao-Yu Guo
Raju Govindaraju
Douglas P Kiel
Margaret Kelly-Hayes
Joseph M Massaro
Michael J Pencina
Sudha Seshadri
Joanne M Murabito
Publikationsdatum
01.09.2007
Verlag
BioMed Central
Erschienen in
BMC Medical Genetics / Ausgabe Sonderheft 1/2007
Elektronische ISSN: 1471-2350
DOI
https://doi.org/10.1186/1471-2350-8-S1-S13

Weitere Artikel der Sonderheft 1/2007

BMC Medical Genetics 1/2007 Zur Ausgabe