1
|
Sebastiani P, Monti S, Lustgarten MS, Song Z, Ellis D, Tian Q, Schwaiger-Haber M, Stancliffe E, Leshchyk A, Short MI, Ardisson Korat AV, Gurinovich A, Karagiannis T, Li M, Lords HJ, Xiang Q, Marron MM, Bae H, Feitosa MF, Wojczynski MK, O'Connell JR, Montasser ME, Schupf N, Arbeev K, Yashin A, Schork N, Christensen K, Andersen SL, Ferrucci L, Rappaport N, Perls TT, Patti GJ. Metabolite signatures of chronological age, aging, survival, and longevity. Cell Rep 2024; 43:114913. [PMID: 39504246 DOI: 10.1016/j.celrep.2024.114913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 07/05/2024] [Accepted: 10/10/2024] [Indexed: 11/08/2024] Open
Abstract
Metabolites that mark aging are not fully known. We analyze 408 plasma metabolites in Long Life Family Study participants to characterize markers of age, aging, extreme longevity, and mortality. We identify 308 metabolites associated with age, 258 metabolites that change over time, 230 metabolites associated with extreme longevity, and 152 metabolites associated with mortality risk. We replicate many associations in independent studies. By summarizing the results into 19 signatures, we differentiate between metabolites that may mark aging-associated compensatory mechanisms from metabolites that mark cumulative damage of aging and from metabolites that characterize extreme longevity. We generate and validate a metabolomic clock that predicts biological age. Network analysis of the age-associated metabolites reveals a critical role of essential fatty acids to connect lipids with other metabolic processes. These results characterize many metabolites involved in aging and point to nutrition as a source of intervention for healthy aging therapeutics.
Collapse
Affiliation(s)
- Paola Sebastiani
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA 02111, USA; Department of Medicine, School of Medicine, Tufts University, Boston, MA 02111, USA.
| | - Stefano Monti
- Department of Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA; Bioinformatics Program, Boston University, Boston, MA 02215, USA
| | - Michael S Lustgarten
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA
| | - Zeyuan Song
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA 02111, USA
| | - Dylan Ellis
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Qu Tian
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD 21224, USA
| | | | - Ethan Stancliffe
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO 63130, USA
| | | | - Meghan I Short
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA 02111, USA; Department of Medicine, School of Medicine, Tufts University, Boston, MA 02111, USA
| | - Andres V Ardisson Korat
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA
| | - Anastasia Gurinovich
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA 02111, USA; Department of Medicine, School of Medicine, Tufts University, Boston, MA 02111, USA
| | - Tanya Karagiannis
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA 02111, USA; Department of Medicine, School of Medicine, Tufts University, Boston, MA 02111, USA
| | - Mengze Li
- Bioinformatics Program, Boston University, Boston, MA 02215, USA
| | - Hannah J Lords
- Bioinformatics Program, Boston University, Boston, MA 02215, USA
| | - Qingyan Xiang
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA 02111, USA
| | - Megan M Marron
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Harold Bae
- Biostatistics Program, College of Health, Oregon State University, Corvallis, OR 97331, USA
| | - Mary F Feitosa
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Mary K Wojczynski
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Jeffrey R O'Connell
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - May E Montasser
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Nicole Schupf
- Department of Epidemiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Konstantin Arbeev
- Social Science Research Institute, Duke University, Durham, NC 27708, USA
| | - Anatoliy Yashin
- Social Science Research Institute, Duke University, Durham, NC 27708, USA
| | - Nicholas Schork
- The Translational Genomics Research Institute, Phoenix, AZ 85004, USA
| | - Kaare Christensen
- Danish Aging Research Center, University of Southern Denmark, 5000 Odense, Denmark
| | - Stacy L Andersen
- Department of Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD 21224, USA
| | - Noa Rappaport
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Thomas T Perls
- Department of Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA
| | - Gary J Patti
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO 63130, USA
| |
Collapse
|
2
|
Yao S, Boudreau RM, Galvin A, Murabito JM, Honig LS, Perls TT, Christensen K, Newman AB. All-Cause Mortality and Cause-Specific Death in U.S. Long-Lived Siblings: Data From the Long Life Family Study. J Gerontol A Biol Sci Med Sci 2024; 79:glae190. [PMID: 39086360 PMCID: PMC11439495 DOI: 10.1093/gerona/glae190] [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: 02/15/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND This study compared the mortality risk of long-lived siblings with the U.S. population average and their spouse controls, and investigated the leading causes of death and the familial effect in death pattern. METHODS In the Long Life Family Study (LLFS), 1 264 proband siblings (mean age 90.1, standard deviation [SD] 6.4) and 172 spouses (83.8, 7.2) from 511 U.S.-based families were recruited and followed more than 12 years. Their survival function was compared with a birth cohort-, baseline age-, sex-, and race-matched pseudo sample from U.S. census data. To examine underlying and contributing causes, we examined in detail 338 deaths with complete death adjudication at the University of Pittsburgh Field Center through the year 2018. A familial effect on survival and death patterns was examined using mixed-effect models. RESULTS The LLFS siblings had better survival than the matched U.S. population average. They also had slightly but not significantly better survival than their spouses' (HR = 1.18 [95%CI 0.94-1.49]) after adjusting for age and sex. Age at death ranged from 75 to 104 years, mean 91.4. The leading causes of death were cardiovascular disease (33.1%), dementia (22.2%), and cancer (10.7%). Mixed effect model shows a significant random effect of family in survival, with adjustment of baseline age and sex. There was no significant familial effect in the underlying cause of death or conditions directly contributing to death among siblings recruited by the University of Pittsburgh Field Center. CONCLUSIONS Our findings demonstrate a higher survival in the LLFS siblings than the U.S. census data, with a familial component of survival. We did not find significant correspondence in causes of death between siblings within families.
Collapse
Affiliation(s)
- Shanshan Yao
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Robert M Boudreau
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Angéline Galvin
- Epidemiology, Biostatistics, and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Joanne M Murabito
- Section of General Internal Medicine, Department of Medicine, and the Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Lawrence S Honig
- Department of Neurology, Sergievsky Center, Taub Institute, Columbia University, New York, New York, USA
| | - Thomas T Perls
- Geriatrics Section, Department of Medicine, Boston Medical Center and Boston University School of Medicine, Boston, Massachusetts, USA
| | - Kaare Christensen
- Epidemiology, Biostatistics, and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Anne B Newman
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
3
|
Feitosa MF, Lin SJ, Acharya S, Thyagarajan B, Wojczynski MK, Kuipers AL, Kulminski A, Christensen K, Zmuda JM, Brent MR, Province MA. Discovery of genomic and transcriptomic pleiotropy between kidney function and soluble receptor for advanced glycation end products using correlated meta-analyses: The Long Life Family Study. Aging Cell 2024; 23:e14261. [PMID: 38932496 PMCID: PMC11464144 DOI: 10.1111/acel.14261] [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: 01/19/2024] [Revised: 06/11/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Patients with chronic kidney disease (CKD) have increased oxidative stress and chronic inflammation, which may escalate the production of advanced glycation end-products (AGEs). High soluble receptor for AGE (sRAGE) and low estimated glomerular filtration rate (eGFR) levels are associated with CKD and aging. We evaluated whether eGFR calculated from creatinine and cystatin C share pleiotropic genetic factors with sRAGE. We employed whole-genome sequencing and correlated meta-analyses on combined genome-wide association study (GWAS) p-values in 4182 individuals (age range: 24-110) from the Long Life Family Study (LLFS). We also conducted transcriptome-wide association studies (TWAS) on whole blood in a subset of 1209 individuals. We identified 59 pleiotropic GWAS loci (p < 5 × 10-8) and 17 TWAS genes (Bonferroni-p < 2.73 × 10-6) for eGFR traits and sRAGE. TWAS genes, LSP1 and MIR23AHG, were associated with eGFR and sRAGE located within GWAS loci, lncRNA-KCNQ1OT1 and CACNA1A/CCDC130, respectively. GWAS variants were eQTLs in the kidney glomeruli and tubules, and GWAS genes predicted kidney carcinoma. TWAS genes harbored eQTLs in the kidney, predicted kidney carcinoma, and connected enhancer-promoter variants with kidney function-related phenotypes at p < 5 × 10-8. Additionally, higher allele frequencies of protective variants for eGFR traits were detected in LLFS than in ALFA-Europeans and TOPMed, suggesting better kidney function in healthy-aging LLFS than in general populations. Integrating genomic annotation and transcriptional gene activity revealed the enrichment of genetic elements in kidney function and aging-related processes. The identified pleiotropic loci and gene expressions for eGFR and sRAGE suggest their underlying shared genetic effects and highlight their roles in kidney- and aging-related signaling pathways.
Collapse
Affiliation(s)
- Mary F. Feitosa
- Division of Statistical Genomics, Department of GeneticsWashington University in St Louis School of MedicineSt. LouisMissouriUSA
| | - Shiow J. Lin
- Division of Statistical Genomics, Department of GeneticsWashington University in St Louis School of MedicineSt. LouisMissouriUSA
| | - Sandeep Acharya
- Department of Computer Science and EngineeringWashington UniversitySt. LouisMissouriUSA
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, School of MedicineUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Mary K. Wojczynski
- Division of Statistical Genomics, Department of GeneticsWashington University in St Louis School of MedicineSt. LouisMissouriUSA
| | - Allison L. Kuipers
- Department of Epidemiology, School of Public HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Alexander Kulminski
- Biodemography of Aging Research Unit, Social Science Research InstituteDuke UniversityDurhamNorth CarolinaUSA
| | - Kaare Christensen
- Unit of Epidemiology, Biostatistics and Biodemography, Department of Public HealthSouthern Denmark UniversityOdenseDenmark
| | - Joseph M. Zmuda
- Department of Epidemiology, School of Public HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Michael R. Brent
- Department of Computer Science and EngineeringWashington UniversitySt. LouisMissouriUSA
| | - Michael A. Province
- Division of Statistical Genomics, Department of GeneticsWashington University in St Louis School of MedicineSt. LouisMissouriUSA
| |
Collapse
|
4
|
Dowrey TW, Cranston SF, Skvir N, Lok Y, Gould B, Petrowitz B, Villar D, Shan J, James M, Dodge M, Belkina AC, Giadone RM, Milman S, Sebastiani P, Perls TT, Andersen SL, Murphy GJ. A longevity-specific bank of induced pluripotent stem cells from centenarians and their offspring. Aging Cell 2024:e14351. [PMID: 39319670 DOI: 10.1111/acel.14351] [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: 04/07/2024] [Revised: 08/02/2024] [Accepted: 09/05/2024] [Indexed: 09/26/2024] Open
Abstract
Centenarians provide a unique lens through which to study longevity, healthy aging, and resiliency. Moreover, models of human aging and resilience to disease that allow for the testing of potential interventions are virtually non-existent. We obtained and characterized over 96 centenarian and offspring peripheral blood samples including those connected to functional independence data highlighting resistance to disability and cognitive impairment. Targeted methylation arrays were used in molecular aging clocks to compare and contrast differences between biological and chronological age in these specialized subjects. Isolated peripheral blood mononuclear cells (PBMCs) from 20 of these subjects were then successfully reprogrammed into high-quality induced pluripotent stem cell (iPSC) lines which were functionally characterized for pluripotency, genomic stability, and the ability to undergo directed differentiation. The result of this work is a one-of-a-kind resource for studies of human longevity and resilience that can fuel the discovery and validation of novel therapeutics for aging-related disease.
Collapse
Affiliation(s)
- Todd W Dowrey
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, Massachusetts, USA
- Section of Hematology and Medical Oncology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Samuel F Cranston
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, Massachusetts, USA
- Section of Hematology and Medical Oncology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Nicholas Skvir
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, Massachusetts, USA
- Section of Hematology and Medical Oncology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Yvonne Lok
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, Massachusetts, USA
- Section of Hematology and Medical Oncology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Brian Gould
- Section of Geriatrics, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Bradley Petrowitz
- Section of Geriatrics, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Daniel Villar
- Albert Einstein College of Medicine, Bronx, New York, USA
| | - Jidong Shan
- Albert Einstein College of Medicine, Bronx, New York, USA
| | - Marianne James
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, Massachusetts, USA
| | - Mark Dodge
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, Massachusetts, USA
| | - Anna C Belkina
- Flow Cytometry Core Facility, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Richard M Giadone
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Sofiya Milman
- Albert Einstein College of Medicine, Bronx, New York, USA
| | - Paola Sebastiani
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, Massachusetts, USA
| | - Thomas T Perls
- Section of Geriatrics, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Stacy L Andersen
- Section of Geriatrics, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - George J Murphy
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, Massachusetts, USA
- Section of Hematology and Medical Oncology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Duan H, Arbeev K, Holmes R, Bagley O, Wu D, Akushevich I, Schupf N, Yashin A, Ukraintseva S. Is being overweight a causal factor in better survival among the oldest old? a Mendelian randomization study. FRONTIERS IN AGING 2024; 5:1442017. [PMID: 39372331 PMCID: PMC11450332 DOI: 10.3389/fragi.2024.1442017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 09/02/2024] [Indexed: 10/08/2024]
Abstract
Overweight, defined by a body mass index (BMI) between 25 and 30, has been associated with enhanced survival among older adults in some studies. However, whether being overweight is causally linked to longevity remains unclear. To investigate this, we conducted a Mendelian randomization (MR) study of lifespan 85+ years, using overweight as an exposure variable and data from the Health and Retirement Study and the Long Life Family Study. An essential aspect of MR involves selecting appropriate single nucleotide polymorphisms (SNPs) as instrumental variables (IVs). This is challenging due to the limited number of SNP candidates within biologically relevant genes that can satisfy all necessary assumptions and criteria. To address this challenge, we employed a novel strategy of creating additional IVs by pairing SNPs between candidate genes. This strategy allowed us to expand the pool of IV candidates with new "composite" SNPs derived from eight candidate obesity genes. Our study found that being overweight between ages 75 and 85, compared to having a normal weight (BMI 18.5-24.9), significantly contributes to improved survival beyond age 85. Results of this MR study thus support a causal relationship between overweight and longevity in older adults.
Collapse
Affiliation(s)
- Hongzhe Duan
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Konstantin Arbeev
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Rachel Holmes
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Olivia Bagley
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Deqing Wu
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Igor Akushevich
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Nicole Schupf
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, United States
| | - Anatoliy Yashin
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Svetlana Ukraintseva
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| |
Collapse
|
6
|
Pflaum JC, Gaertner VD, Brandstetter S, Apfelbacher C, Melter M, Koeninger A, Kabesch M. Defining familial longevity and developing a familial longevity score for unbiased epigenetic studies in a birth cohort. Epigenomics 2024; 16:1149-1158. [PMID: 39264702 PMCID: PMC11457659 DOI: 10.1080/17501911.2024.2370760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 06/18/2024] [Indexed: 09/13/2024] Open
Abstract
Aim: Longevity accumulating in families has genetic and epigenetic components. To study early and unbiased epigenetic predictors of longevity prospectively, a birth cohort would be ideal. However, the original family longevity selection score (FLoSS) focuses on populations of elderly only.Methods: In the German birth cohort KUNO-Kids we assessed when information for such scores may be best collected and how to calculate an adapted FLoSS.Results: A total of 551 families contributed to adapted FLoSS, with a mean score of -0.15 (SD 2.33). Adapted FLoSS ≥7 as a marker of exceptional longevity occurred in 3.3% of families, comparable to original FLoSS in elderly.Conclusion: An adapted FLoSS from data collectable postnatally may be a feasible tool to study unbiased epigenetic predictors for longevity.
Collapse
Affiliation(s)
- Jasmin C Pflaum
- Department of Pediatric Pneumology & Allergy, University Children's Hospital Regensburg (KUNO), of the University of Regensburg & the Order of St. John at the St. Hedwig Hospital, Regensburg, Germany
| | - Vincent D Gaertner
- Department of Pediatric Pneumology & Allergy, University Children's Hospital Regensburg (KUNO), of the University of Regensburg & the Order of St. John at the St. Hedwig Hospital, Regensburg, Germany
| | - Susanne Brandstetter
- Science & Innovation Campus Regensburg (WECARE) of the Order of St. John at the St. Hedwig Hospital, Regensburg, Germany
- University Children's Hospital Regensburg (KUNO) of the University of Regensburg & the Order of St. John at the St. Hedwig Hospital, Regensburg, Germany
| | - Christian Apfelbacher
- Science & Innovation Campus Regensburg (WECARE) of the Order of St. John at the St. Hedwig Hospital, Regensburg, Germany
- Institute of Social Medicine & Health Systems Research, Otto von Guericke University, Magdeburg, Germany
| | - Michael Melter
- University Children's Hospital Regensburg (KUNO) of the University of Regensburg & the Order of St. John at the St. Hedwig Hospital, Regensburg, Germany
| | - Angela Koeninger
- Department of Obstetrics & Gynaecology of the University of Regensburg & the Order of St. John at the St. Hedwig Hospital, Regensburg, Germany
| | - Michael Kabesch
- Department of Pediatric Pneumology & Allergy, University Children's Hospital Regensburg (KUNO), of the University of Regensburg & the Order of St. John at the St. Hedwig Hospital, Regensburg, Germany
- Science & Innovation Campus Regensburg (WECARE) of the Order of St. John at the St. Hedwig Hospital, Regensburg, Germany
| |
Collapse
|
7
|
Santanasto AJ, Acharya S, Wojczynski MK, Cvejkus RK, Lin S, Brent MR, Anema JA, Wang L, Thyagarajan B, Christensen K, Daw EW, Zmuda JM. Whole Genome Linkage and Association Analyses Identify DLG Associated Protein-1 as a Novel Positional and Biological Candidate Gene for Muscle Strength: The Long Life Family Study. J Gerontol A Biol Sci Med Sci 2024; 79:glae144. [PMID: 38808484 PMCID: PMC11226997 DOI: 10.1093/gerona/glae144] [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: 09/20/2023] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND Grip strength is a robust indicator of overall health, is moderately heritable, and predicts longevity in older adults. METHODS Using genome-wide linkage analysis, we identified a novel locus on chromosome 18p (mega-basepair region: 3.4-4.0) linked to grip strength in 3 755 individuals from 582 families aged 64 ± 12 years (range 30-110 years; 55% women). There were 26 families that contributed to the linkage peak (cumulative logarithm of the odds [LOD] score = 10.94), with 6 families (119 individuals) accounting for most of the linkage signal (LOD = 6.4). In these 6 families, using whole genome sequencing data, we performed association analyses between the 7 312 single nucleotide (SNVs) and insertion deletion (INDELs) variants in the linkage region and grip strength. Models were adjusted for age, age2, sex, height, field center, and population substructure. RESULTS We found significant associations between genetic variants (8 SNVs and 4 INDELs, p < 5 × 10-5) in the Disks Large-associated Protein 1 (DLGAP1) gene and grip strength. Haplotypes constructed using these variants explained up to 98.1% of the LOD score. Finally, RNAseq data showed that these variants were significantly associated with the expression of nearby Myosin Light Chain 12A (MYL12A), Structural Maintenance of Chromosomes Flexible Hinge Domain Containing 1 (SMCHD1), Erythrocyte Membrane Protein Band 4.1 Like 3 (EPB41L3) genes (p < .0004). CONCLUSIONS The DLGAP1 gene plays an important role in the postsynaptic density of neurons; thus, it is both a novel positional and biological candidate gene for follow-up studies aimed at uncovering genetic determinants of muscle strength.
Collapse
Affiliation(s)
- Adam J Santanasto
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sandeep Acharya
- Division of Computational and Data Sciences, Center for Genome Sciences and Systems Biology, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Computer Science, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Mary K Wojczynski
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Ryan K Cvejkus
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Shiow Lin
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Michael R Brent
- Division of Computational and Data Sciences, Center for Genome Sciences and Systems Biology, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Computer Science, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Jason A Anema
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Lihua Wang
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kaare Christensen
- Epidemiology Unit, Institute of Public Health, The Danish Aging Research Center, University of Southern Denmark, Odense, Denmark
| | - E Warwick Daw
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Joseph M Zmuda
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
8
|
Arbeev KG, Bagley O, Ukraintseva SV, Kulminski A, Stallard E, Schwaiger-Haber M, Patti GJ, Gu Y, Yashin AI, Province MA. Methods for joint modelling of longitudinal omics data and time-to-event outcomes: Applications to lysophosphatidylcholines in connection to aging and mortality in the Long Life Family Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.29.24311176. [PMID: 39132492 PMCID: PMC11312646 DOI: 10.1101/2024.07.29.24311176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Studying relationships between longitudinal changes in omics variables and risks of events requires specific methodologies for joint analyses of longitudinal and time-to-event outcomes. We applied two such approaches (joint models [JM], stochastic process models [SPM]) to longitudinal metabolomics data from the Long Life Family Study focusing on understudied associations of longitudinal changes in lysophosphatidylcholines (LPC) with mortality and aging-related outcomes (23 LPC species, 5,790 measurements of each in 4,011 participants, 1,431 of whom died during follow-up). JM analyses found that higher levels of the majority of LPC species were associated with lower mortality risks, with the largest effect size observed for LPC 15:0/0:0 (hazard ratio: 0.715, 95% CI (0.649, 0.788)). SPM applications to LPC 15:0/0:0 revealed how the association found in JM reflects underlying aging-related processes: decline in robustness to deviations from optimal LPC levels, better ability of males' organisms to return to equilibrium LPC levels (which are higher in females), and increasing gaps between the optimum and equilibrium levels leading to increased mortality risks with age. Our results support LPC as a biomarker of aging and related decline in robustness/resilience, and call for further exploration of factors underlying age-dynamics of LPC in relation to mortality and diseases.
Collapse
Affiliation(s)
- Konstantin G. Arbeev
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina 27708, USA
| | - Olivia Bagley
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina 27708, USA
| | - Svetlana V. Ukraintseva
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina 27708, USA
| | - Alexander Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina 27708, USA
| | - Eric Stallard
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina 27708, USA
| | - Michaela Schwaiger-Haber
- Department of Chemistry, Washington University in St. Louis, St. Louis, Missouri 63130, United States
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri 63130, United States
- Center for Metabolomics and Isotope Tracing at Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Gary J. Patti
- Department of Chemistry, Washington University in St. Louis, St. Louis, Missouri 63130, United States
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri 63130, United States
- Center for Metabolomics and Isotope Tracing at Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Yian Gu
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
- G.H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, New York, New York 10032, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York 10032, USA
| | - Anatoliy I. Yashin
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina 27708, USA
| | - Michael A. Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| |
Collapse
|
9
|
Duan H, Arbeev K, Holmes R, Bagley O, Wu D, Akushevich I, Schupf N, Yashin A, Ukraintseva S. Overweight as a Causal Factor Contributing to Better Survival at the Oldest Old Ages: A Mendelian Randomization Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.30.24308211. [PMID: 38853995 PMCID: PMC11160847 DOI: 10.1101/2024.05.30.24308211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Overweight, defined by a body mass index (BMI) between 25 and 30, has been associated with enhanced survival among older adults in some studies. However, whether being overweight is causally linked to longevity remains unclear. To investigate this, we conducted a Mendelian randomization (MR) study of lifespan 85+ years, using overweight as an exposure variable and data from the Health and Retirement Study and the Long Life Family Study. An essential aspect of MR involves selecting appropriate single-nucleotide polymorphisms (SNPs) as instrumental variables (IVs). This is challenging due to the limited number of SNP candidates within biologically relevant genes that can satisfy all necessary assumptions and criteria. To address this challenge, we employed a novel strategy of creating additional IVs by pairing SNPs between candidate genes. This strategy allowed us to expand the pool of IV candidates with new 'composite' SNPs derived from eight candidate obesity genes. Our study found that being overweight between ages 75 and 85, compared to having a normal weight (BMI 18.5-24.9), significantly contributes to improved survival beyond age 85. Results of this MR study thus support a causal relationship between overweight and longevity in older adults.
Collapse
Affiliation(s)
- Hongzhe Duan
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Konstantin Arbeev
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Rachel Holmes
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Olivia Bagley
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Deqing Wu
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Igor Akushevich
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Nicole Schupf
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York, United States
| | - Anatoliy Yashin
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Svetlana Ukraintseva
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| |
Collapse
|
10
|
Daw EW, Anema JA, Schwander K, Lin SJ, Wang L, Wojczynski M, Thyagarajan B, Stitziel N, Province MA. A Paradigm For Calling Sequence In Families: The Long Life Family Study. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.23.595584. [PMID: 38826248 PMCID: PMC11142154 DOI: 10.1101/2024.05.23.595584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Over Several years, we have developed a system for assuring the quality of whole genome sequence (WGS) data in the LLFS families. We have focused on providing data to identify germline genetic variants with the aim of releasing as many variants on as many individuals as possible. We aim to assure the quality of the individual calls. The availability of family data has enabled us to use and validate some filters not commonly used in population-based studies. We developed slightly different procedures for the autosomal, X, Y, and Mitochondrial (MT) chromosomes. Some of these filters are specific to family data, but some can be used with any WGS data set. We also describe the procedure we use to construct linkage markers from the SNP sequence data and how we compute IBD values for use in linkage analysis.
Collapse
|
11
|
Patel R, Cosentino S, Zheng EZ, Schupf N, Barral S, Feitosa M, Andersen SL, Sebastiani P, Ukraintseva S, Christensen K, Zmuda J, Thyagarajan B, Gu Y. Systemic inflammation in relation to exceptional memory in the Long Life Family Study (LLFS). Brain Behav Immun Health 2024; 37:100746. [PMID: 38476338 PMCID: PMC10925922 DOI: 10.1016/j.bbih.2024.100746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 02/12/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Background and objectives We previously found a substantial familial aggregation of healthy aging phenotypes, including exceptional memory (EM) in long-lived persons. In the current study, we aim to assess whether long-lived families with EM and without EM (non-EM) differ in systemic inflammation status and trajectory. Methods The current study included 4333 participants of the multi-center Long Life Family Study (LLFS). LLFS families were classified as EM (556 individuals from 28 families) or non-EM (3777 individuals from 416 families), with 2 or more offspring exhibiting exceptional memory performance (i.e. having baseline composite z-score representing immediate and delayed story memory being 1.5 SD above the mean in the nondemented offspring sample) considered as EM. Blood samples from baseline were used to measure inflammatory biomarkers including total white blood cell (WBC) and its subtypes (neutrophils, lymphocytes, monocytes) count, platelet count, high sensitivity C-reactive protein, and interleukin-6. Generalized linear models were used to examine cross-sectional differences in inflammatory biomarkers at baseline. In a sub-sample of 2227 participants (338 subjects from 24 EM families and 1889 from 328 non-EM families) with repeated measures of immune cell counts, we examined whether the rate of biomarker change differed between EM and non-EM families. All models were adjusted for family size, relatedness, age, sex, education, field center, APOE genotype, and body mass index. Results LLFS participants from EM families had a marginally higher monocyte count at baseline (b = 0.028, SE = 0.0110, p = 0.010) after adjusting for age, sex, education, and field site, particularly in men (p < 0.0001) but not in women (p = 0.493) (p-interaction = 0.003). Over time, monocyte counts increased (p < 0.0001) in both EM and non-EM families, while lymphocytes and platelet counts decreased over time in the non-EM families (p < 0.0001) but not in the EM families. After adjusting for multiple variables, there was no significant difference in biomarker change over time between the EM and non-EM families. Discussion Compared with non-EM families, EM families had significantly higher monocyte count at baseline but had similar change over time. Our study suggests that differences in monocyte counts may be a pathway through which EM emerges in some long-lived families, especially among men.
Collapse
Affiliation(s)
- Ruhee Patel
- Cognitive Neuroscience Division, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Stephanie Cosentino
- Cognitive Neuroscience Division, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Esther Zhiwei Zheng
- Cognitive Neuroscience Division, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Nicole Schupf
- Cognitive Neuroscience Division, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Sandra Barral
- Cognitive Neuroscience Division, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Mary Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Stacy L. Andersen
- Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Paola Sebastiani
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA, 02111, USA
| | | | - Kaare Christensen
- Epidemiology, Biostatistics and Biodemography, University of Southern Denmark, 5230, Odense, Denmark
| | - Joseph Zmuda
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Yian Gu
- Cognitive Neuroscience Division, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Long Life Family Study (LLFS)
- Cognitive Neuroscience Division, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA, 02111, USA
- Social Sciences Research Institute, Duke University, Durham, NC, 27705, USA
- Epidemiology, Biostatistics and Biodemography, University of Southern Denmark, 5230, Odense, Denmark
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA
| |
Collapse
|
12
|
Song Z, Gunn S, Monti S, Peloso GM, Liu CT, Lunetta K, Sebastiani P. Learning Gaussian Graphical Models from Correlated Data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.03.587948. [PMID: 38617340 PMCID: PMC11014549 DOI: 10.1101/2024.04.03.587948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Gaussian Graphical Models (GGM) have been widely used in biomedical research to explore complex relationships between many variables. There are well established procedures to build GGMs from a sample of independent and identical distributed observations. However, many studies include clustered and longitudinal data that result in correlated observations and ignoring this correlation among observations can lead to inflated Type I error. In this paper, we propose a Bootstrap algorithm to infer GGM from correlated data. We use extensive simulations of correlated data from family-based studies to show that the Bootstrap method does not inflate the Type I error while retaining statistical power compared to alternative solutions. We apply our method to learn the GGM that represents complex relations between 47 Polygenic Risk Scores generated using genome-wide genotype data from a family-based study known as the Long Life Family Study. By comparing it to the conventional methods that ignore within-cluster correlation, we show that our method controls the Type I error well in this real example.
Collapse
|
13
|
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
|
14
|
Dowrey TW, Cranston SF, Skvir N, Lok Y, Gould B, Petrowitz B, Villar D, Shan J, James M, Dodge M, Belkina AC, Giadone RM, Sebastiani P, Perls TT, Andersen SL, Murphy GJ. A longevity-specific bank of induced pluripotent stem cells from centenarians and their offspring. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.12.584663. [PMID: 38559230 PMCID: PMC10979955 DOI: 10.1101/2024.03.12.584663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Centenarians provide a unique lens through which to study longevity, healthy aging, and resiliency. Moreover, models of human aging and resilience to disease that allow for the testing of potential interventions are virtually non-existent. We obtained and characterized over 50 centenarian and offspring peripheral blood samples including those connected to functional independence data highlighting resistance to disability and cognitive impairment. Targeted methylation arrays were used in molecular aging clocks to compare and contrast differences between biological and chronological age in these specialized subjects. Isolated peripheral blood mononuclear cells (PBMCs) were then successfully reprogrammed into high-quality induced pluripotent stem cell (iPSC) lines which were functionally characterized for pluripotency, genomic stability, and the ability to undergo directed differentiation. The result of this work is a one-of-a-kind resource for studies of human longevity and resilience that can fuel the discovery and validation of novel therapeutics for aging-related disease.
Collapse
|
15
|
Galvin A, Pedersen JK, Wojczynski MK, Ukraintseva S, Arbeev K, Feitosa M, Province MA, Christensen K. The Protective Effect of Familial Longevity Persists After Age 100: Findings From the Danish National Registers. J Gerontol A Biol Sci Med Sci 2024; 79:glad164. [PMID: 37449765 PMCID: PMC10733167 DOI: 10.1093/gerona/glad164] [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: 01/26/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND A recent study suggested that the protective effect of familial longevity becomes negligible for centenarians. However, the authors assessed the dependence on familial longevity in centenarians by comparing centenarians with 1 parent surviving to age 80+ to centenarians whose same-sexed parent did not survive to age 80. Here we test whether the protective effect of familial longevity persists after age 100 using more restrictive definitions of long-lived families. METHODS Long-lived sibships were identified through 3 nationwide, consecutive studies in Denmark, including families with either at least 2 siblings aged 90+ or a Family Longevity Selection Score (FLoSS) above 7. Long-lived siblings enrolled in these studies and who reached age 100 were included. For each sibling, 5 controls matched on sex and year of birth were randomly selected among centenarians in the Danish population. Survival time from age 100 was described with Kaplan-Meier curves for siblings and controls separately. Survival analyses were performed using stratified Cox proportional hazards models. RESULTS A total of 340 individuals from long-lived sibships who survived to age 100 and 1 700 controls were included. Among the long-lived siblings and controls, 1 650 (81%) were women. The results showed that long-lived siblings presented better overall survival after age 100 than sporadic long-livers (hazard ratio [HR] = 0.80, 95% confidence interval [CI] = 0.71-0.91), with even lower estimate (HR = 0.65, 95% CI = 0.50-0.85) if familial longevity was defined by FLoSS. CONCLUSIONS The present study, with virtually no loss to follow-up, demonstrated a persistence of protective effect of familial longevity after age 100.
Collapse
Affiliation(s)
- Angéline Galvin
- Epidemiology, Biostatistics, and Biodemography Team, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Jacob Krabbe Pedersen
- Epidemiology, Biostatistics, and Biodemography Team, Department of Public Health, University of Southern Denmark, Odense, Denmark
- The Danish Aging Research Center, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Mary K Wojczynski
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Svetlana Ukraintseva
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina, USA
| | - Konstantin Arbeev
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina, USA
| | - Mary Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michael A Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Kaare Christensen
- Epidemiology, Biostatistics, and Biodemography Team, Department of Public Health, University of Southern Denmark, Odense, Denmark
- The Danish Aging Research Center, Department of Public Health, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
16
|
Galvin A, Pedersen JK, Arbeev KG, Feitosa MF, Ukraintseva S, Yao S, Newman AB, Christensen K. End of Life Events and Causes of Death in Danish Long-Lived Siblings: Reduced Dementia Risk Compared to Sporadic Long-Livers. J Alzheimers Dis 2024; 99:1397-1407. [PMID: 38788069 DOI: 10.3233/jad-231204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
Background Better physical robustness and resilience of long-lived siblings compared to sporadic long-livers has been demonstrated in several studies. However, it is unknown whether long-lived siblings also end their lives better. Objective To investigate end-of-life (EoL) events (dementia diagnosis, medication, hospitalizations in the last 5 years of life), causes of death, and location of death in long-lived siblings compared to matched sporadic long-livers from the Danish population. Methods Long-lived siblings were identified through three nationwide Danish studies in which the inclusion criteria varied, but 99.5% of the families had at least two siblings surviving to age 90 + . Those who died between 2006 and 2018 were included, and randomly matched with sex, year-of-birth and age-at-death controls (i.e., sporadic long-lived controls) from the Danish population. Results A total of 5,262 long-lived individuals were included (1,754 long-lived siblings, 3,508 controls; 63% women; median age at death 96.1). Long-lived siblings had a significantly lower risk of being diagnosed with dementia in the last years of life (p = 0.027). There was no significant difference regarding the number of prescribed drugs, hospital stays, days in hospital, and location of death. Compared to controls, long-lived siblings presented a lower risk of dying from dementia (p = 0.020) and ill-defined conditions (p = 0.030). Conclusions In many aspects long-lived siblings end their lives similar to sporadic long-livers, with the important exception of lower dementia risk during the last 5 years of life. These results suggest that long-lived siblings are excellent candidates for identifying environmental and genetic protective factors of dementia.
Collapse
Affiliation(s)
- Angéline Galvin
- Epidemiology, Biostatistics, and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Jacob Krabbe Pedersen
- Epidemiology, Biostatistics, and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
- Department of Public Health, The Danish Aging Research Center, University of Southern Denmark, Odense, Denmark
| | - Konstantin G Arbeev
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, USA
| | - Mary F Feitosa
- Department of Genetics, Division of Statistical Genomics, Washington University School of Medicine, St. Louis, MO, USA
| | - Svetlana Ukraintseva
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, USA
| | - Shanshan Yao
- Department of Epidemiology, Center for Aging and Population Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anne B Newman
- Department of Epidemiology, Center for Aging and Population Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Geriatric Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kaare Christensen
- Epidemiology, Biostatistics, and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
- Department of Public Health, The Danish Aging Research Center, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
17
|
Schumacher BT, Kehler DS, Kulminski AM, Qiao Y(S, Andersen SL, Gmelin T, Christensen K, Wojczynski MK, Theou O, Rockwood K, Newman AB, Glynn NW. The association between frailty and perceived physical and mental fatigability: The Long Life Family Study. J Am Geriatr Soc 2024; 72:219-225. [PMID: 37814920 PMCID: PMC10843058 DOI: 10.1111/jgs.18624] [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: 05/12/2023] [Revised: 08/21/2023] [Accepted: 09/11/2023] [Indexed: 10/11/2023]
Abstract
BACKGROUND Higher levels of frailty, quantified by a frailty index (FI), may be linked to fatigue severity as tasks become more physically and mentally demanding. Fatigue, a component of frailty research, has been ambiguous and inconsistent in its operationalization. Fatigability-the quantification of vulnerability to fatigue in relation to specific intensity and duration of activities-offers a more sensitive and standardized approach, though the association between frailty and fatigability has not been assessed. METHODS Using cross-sectional data from the Long Life Family Study at Visit 2 (2014-2017; N = 2524; mean age ± standard deviation (SD) 71.4 ± 11.2 years; 55% women; 99% White), we examined associations between an 83-item FI after excluding fatigue items (ratio of number of health problems reported (numerator) out of the total assessed (denominator); higher ratio = greater frailty) and perceived physical and mental fatigability using the Pittsburgh Fatigability Scale (PFS) (range 0-50; higher scores = greater fatigability). RESULTS Participants had mean ± standard deviation FI (0.08 ± 0.06; observed range: 0.0-0.43), PFS Physical (13.7 ± 9.6; 39.5% more severe, ≥15), and PFS Mental (7.9 ± 8.9; 22.8% more severe, ≥13). The prevalence of more severe physical and mental fatigability was higher across FI quartiles. In mixed effects models accounting for family structure, a clinically meaningful 3%-higher FI was associated with 1.9 points higher PFS Physical score (95% confidence interval (CI) 1.7-2.1) and 1.7 points higher PFS Mental score (95% CI 1.5-1.9) after adjusting for covariates. CONCLUSIONS Frailty was associated with perceived physical and mental fatigability severity. Understanding this association may support the development of interventions to mitigate the risks associated with greater frailty and perceived fatigability. Including measurements of perceived fatigability, in lieu of fatigue, in frailty indices has the potential to alleviate the inconsistencies and ambiguity surrounding the operationalization of fatigue and provide a more precise and sensitive measurement of frailty.
Collapse
Affiliation(s)
| | - Dustin S. Kehler
- School of Physiotherapy, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Alexander M. Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, USA
| | - Yujia (Susanna) Qiao
- Department of Epidemiology, School of Public Health, University of Pittsburgh, PA, USA
| | - Stacy L. Andersen
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Theresa Gmelin
- Department of Epidemiology, School of Public Health, University of Pittsburgh, PA, USA
| | - Kaare Christensen
- Danish Aging Research Center, University of Southern Denmark
- Department of Clinical Biochemistry and Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | | | - Olga Theou
- School of Physiotherapy, Dalhousie University, Halifax, Nova Scotia, Canada
- Division of Geriatric Medicine, Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kenneth Rockwood
- Division of Geriatric Medicine, Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Anne B. Newman
- Department of Epidemiology, School of Public Health, University of Pittsburgh, PA, USA
| | - Nancy W. Glynn
- Department of Epidemiology, School of Public Health, University of Pittsburgh, PA, USA
| | | |
Collapse
|
18
|
Arbeev KG, Ukraintseva S, Bagley O, Duan H, Wu D, Akushevich I, Stallard E, Kulminski A, Christensen K, Feitosa MF, O’Connell JR, Parker D, Whitson H, Yashin AI. Interactions between genes involved in physiological dysregulation and axon guidance: role in Alzheimer's disease. Front Genet 2023; 14:1236509. [PMID: 37719713 PMCID: PMC10500346 DOI: 10.3389/fgene.2023.1236509] [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: 06/07/2023] [Accepted: 08/17/2023] [Indexed: 09/19/2023] Open
Abstract
Dysregulation of physiological processes may contribute to Alzheimer's disease (AD) development. We previously found that an increase in the level of physiological dysregulation (PD) in the aging body is associated with declining resilience and robustness to major diseases. Also, our genome-wide association study found that genes associated with the age-related increase in PD frequently represented pathways implicated in axon guidance and synaptic function, which in turn were linked to AD and related traits (e.g., amyloid, tau, neurodegeneration) in the literature. Here, we tested the hypothesis that genes involved in PD and axon guidance/synapse function may jointly influence onset of AD. We assessed the impact of interactions between SNPs in such genes on AD onset in the Long Life Family Study and sought to replicate the findings in the Health and Retirement Study. We found significant interactions between SNPs in the UNC5C and CNTN6, and PLXNA4 and EPHB2 genes that influenced AD onset in both datasets. Associations with individual SNPs were not statistically significant. Our findings, thus, support a major role of genetic interactions in the heterogeneity of AD and suggest the joint contribution of genes involved in PD and axon guidance/synapse function (essential for the maintenance of complex neural networks) to AD development.
Collapse
Affiliation(s)
- Konstantin G. Arbeev
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Svetlana Ukraintseva
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Olivia Bagley
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Hongzhe Duan
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Deqing Wu
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Igor Akushevich
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Eric Stallard
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Alexander Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Kaare Christensen
- Danish Aging Research Center, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Mary F. Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, United States
| | - Jeffrey R. O’Connell
- Division of Endocrinology, Diabetes and Nutrition and Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Daniel Parker
- Duke Center for the Study of Aging and Human Development, Duke University, Durham, NC, United States
| | - Heather Whitson
- Duke Center for the Study of Aging and Human Development, Duke University, Durham, NC, United States
- Durham VA Geriatrics Research Education and Clinical Center, Durham, NC, United States
| | - Anatoliy I. Yashin
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| |
Collapse
|
19
|
Song Z, Gurinovich A, Nygaard M, Mengel-From J, Andersen S, Cosentino S, Schupf N, Lee J, Zmuda J, Ukraintseva S, Arbeev K, Christensen K, Perls T, Sebastiani P. Rare genetic variants correlate with better processing speed. Neurobiol Aging 2023; 125:115-122. [PMID: 36813607 PMCID: PMC10038891 DOI: 10.1016/j.neurobiolaging.2022.11.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/17/2022] [Accepted: 11/23/2022] [Indexed: 01/30/2023]
Abstract
We conducted a genome-wide association study of Digit Symbol Substitution Test scores administered in 4207 family members of the Long Life Family Study (LLFS). Genotype data were imputed to the HRC panel of 64,940 haplotypes resulting in ∼15M genetic variants with a quality score > 0.7. The results were replicated using genetic data imputed to the 1000 Genomes phase 3 reference panel from 2 Danish twin cohorts: the study of Middle Aged Danish Twins and the Longitudinal Study of Aging Danish Twins. The genome-wide association study in LLFS discovered 18 rare genetic variants (minor allele frequency (MAF) < 1.0%) that reached genome-wide significance (p-value < 5 × 10-8). Among these, 17 rare variants in chromosome 3 had large protective effects on the processing speed, including rs7623455, rs9821776, rs9821587, rs78704059, which were replicated in the combined Danish twin cohort. These SNPs are located in/near 2 genes, THRB and RARB, that belonged to the thyroid hormone receptors family that may influence the speed of metabolism and cognitive aging. The gene-level tests in LLFS confirmed that these 2 genes are associated with processing speed.
Collapse
Affiliation(s)
- Zeyuan Song
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA.
| | - Anastasia Gurinovich
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA, USA
| | - Marianne Nygaard
- Epidemiology, Biostatistics and Biodemography, The Danish Aging Research Center, and The Danish Twin Registry, Institute of Public Health, University of Southern Denmark, Odense, Denmark; Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Jonas Mengel-From
- Epidemiology, Biostatistics and Biodemography, The Danish Aging Research Center, and The Danish Twin Registry, Institute of Public Health, University of Southern Denmark, Odense, Denmark; Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Stacy Andersen
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Stephanie Cosentino
- Departments of Epidemiology and Neurology, Columbia University Medical Center, New York, NY, USA
| | - Nicole Schupf
- Departments of Epidemiology and Neurology, Columbia University Medical Center, New York, NY, USA
| | - Joseph Lee
- Departments of Epidemiology and Neurology, Columbia University Medical Center, New York, NY, USA
| | - Joseph Zmuda
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Svetlana Ukraintseva
- 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
| | - Kaare Christensen
- Epidemiology, Biostatistics and Biodemography, The Danish Aging Research Center, and The Danish Twin Registry, Institute of Public Health, University of Southern Denmark, Odense, Denmark; Department of Clinical Genetics, Odense University Hospital, Odense, Denmark; Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - Thomas Perls
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Paola Sebastiani
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA, USA
| |
Collapse
|
20
|
Loika Y, Loiko E, Feng F, Stallard E, Yashin AI, Arbeev K, Kuipers AL, Feitosa MF, Province MA, Kulminski AM. Exogenous exposures shape genetic predisposition to lipids, Alzheimer's, and coronary heart disease in the MLXIPL gene locus. Aging (Albany NY) 2023; 15:3249-3272. [PMID: 37074818 PMCID: PMC10449285 DOI: 10.18632/aging.204665] [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: 10/13/2022] [Accepted: 04/06/2023] [Indexed: 04/20/2023]
Abstract
Associations of single nucleotide polymorphisms (SNPs) of the MLXIPL lipid gene with Alzheimer's (AD) and coronary heart disease (CHD) and potentially causal mediation effects of their risk factors, high-density lipoprotein cholesterol (HDL-C) and triglycerides (TG), were examined in two samples of European ancestry from the US (22,712 individuals 587/2,608 AD/CHD cases) and the UK Biobank (UKB) (232,341 individuals; 809/15,269 AD/CHD cases). Our results suggest that these associations can be regulated by several biological mechanisms and shaped by exogenous exposures. Two patterns of associations (represented by rs17145750 and rs6967028) were identified. Minor alleles of rs17145750 and rs6967028 demonstrated primary (secondary) association with high TG (lower HDL-C) and high HDL-C (lower TG) levels, respectively. The primary association explained ~50% of the secondary one suggesting partly independent mechanisms of TG and HDL-C regulation. The magnitude of the association of rs17145750 with HDL-C was significantly higher in the US vs. UKB sample and likely related to differences in exogenous exposures in the two countries. rs17145750 demonstrated a significant detrimental indirect effect through TG on AD risk in the UKB only (βIE = 0.015, pIE = 1.9 × 10-3), which suggests protective effects of high TG levels against AD, likely shaped by exogenous exposures. Also, rs17145750 demonstrated significant protective indirect effects through TG and HDL-C in the associations with CHD in both samples. In contrast, rs6967028 demonstrated an adverse mediation effect through HDL-C on CHD risk in the US sample only (βIE = 0.019, pIE = 8.6 × 10-4). This trade-off suggests different roles of triglyceride mediated mechanisms in the pathogenesis of AD and CHD.
Collapse
Affiliation(s)
- Yury Loika
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC 27708, USA
| | - Elena Loiko
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC 27708, USA
| | - Fan Feng
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC 27708, USA
| | - Eric Stallard
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC 27708, USA
| | - Anatoliy I. Yashin
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC 27708, USA
| | - Konstantin Arbeev
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC 27708, USA
| | - Allison L. Kuipers
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Mary F. Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Michael A. Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Alexander M. Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC 27708, USA
| |
Collapse
|
21
|
Katz R, Gay EL, Kuipers AL, Lee JH, Honig LS, Christensen K, Feitosa MF, Wojczynski MK, Glynn NW. Association of leukocyte telomere length with perceived physical fatigability. Exp Gerontol 2022; 170:111988. [PMID: 36302456 PMCID: PMC11467958 DOI: 10.1016/j.exger.2022.111988] [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: 03/26/2022] [Revised: 09/22/2022] [Accepted: 10/19/2022] [Indexed: 12/29/2022]
Abstract
BACKGROUND Leukocyte telomere length (LTL) is a potential genomic marker of biological aging, but its relation to fatigability, a prognostic indicator of phenotypic aging (e.g., functional decline) is unknown. We hypothesized shorter LTL would predict greater perceived physical fatigability, but that this association would be attenuated by adjusting for chronological age. METHODS Two generations of participants (N = 1997; 309 probands, 1688 offspring) were from the Long Life Family Study (age = 73.7 ± 10.4, range 60-108, 54.4 % women), a longitudinal cohort study of aging. LTL was assayed at baseline. Perceived physical fatigability was measured 8.0 ± 1.1 years later using the validated, self-administered 10-item Pittsburgh Fatigability Scale (PFS, 0-50, higher scores = greater fatigability). Generalized estimating equations were generated to model the association between LTL and PFS Physical scores. RESULTS Prevalence of greater physical fatigability (PFS scores≥15) was 41.9 %. Using generalized estimating equations, a one kilobase pair shorter LTL was associated with higher PFS Physical scores (β = 1.8, p < .0001), accounting for family structure, and adjusting for field center, follow-up time, sex, and follow-up body mass index, physical activity, and chronic health conditions. When age was included as a covariate, the association was fully attenuated (β = 0.1, p = .78). CONCLUSION LTL may provide an alternative method for estimating an individual's lifetime exposure to chronic stressors, but does not appear to provide additional information not captured by chronological age. Further research is needed to characterize the interaction between age, LTL, and perceived fatigability, and develop a method of identifying individuals at risk for deleterious aging.
Collapse
Affiliation(s)
- Rain Katz
- School of Public Health, Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Emma L Gay
- School of Public Health, Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Allison L Kuipers
- School of Public Health, Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Joseph H Lee
- Sergievsky Center, Taub Institute, Department of Epidemiology, Columbia University, New York, NY, USA; Sergievsky Center, Taub Institute, Department of Neurology, Columbia University, New York, NY, USA
| | - Lawrence S Honig
- Sergievsky Center, Taub Institute, Department of Neurology, Columbia University, New York, NY, USA
| | - Kaare Christensen
- Department of Public Health, Unit of Epidemiology, Biostatistics and Biodemography, University of Southern Denmark, Odense, Denmark
| | - Mary F Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Mary K Wojczynski
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Nancy W Glynn
- School of Public Health, Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
22
|
Galvin A, Feitosa M, Arbeev K, Kuipers AL, Wojczynski M, Ukrainsteva S, Christensen K. Physical resilience after a diagnosis of cardiovascular disease among offspring of long-lived siblings. Eur J Ageing 2022; 19:437-445. [PMID: 36052181 PMCID: PMC9424427 DOI: 10.1007/s10433-021-00641-7] [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] [Accepted: 07/12/2021] [Indexed: 11/27/2022] Open
Abstract
Health benefits of longevity-enriched families transmit across generations and a lower incidence of cardiovascular diseases (CVD) have been shown to contribute to this phenomenon. In the current study, we investigated whether the offspring of long-lived siblings also have better survival after a CVD diagnosis compared to matched controls, i.e., are they both robust and resilient? Offspring of long-lived siblings were identified from three nationwide Danish studies and linked to national registers. Offspring with first diagnosis of acute myocardial infarction, chronic ischemic heart disease, heart failure or cerebrovascular disease between 1996 and 2011 were included and matched with two controls from the Danish population on sex, year of birth and diagnosis, and type of CVD. Stratified Cox proportional-hazards models on the matching data were performed to study 10-year overall survival. A total of 402 offspring and 804 controls were included: 64.2% male with a median age at diagnosis of 63.0. For offspring and controls, overall survival was 73% and 65% at 10 years from diagnosis, respectively. Offspring of long-lived siblings had a significantly better survival than controls, and this association was slightly attenuated after controlling for marital status, medication and Charlson Comorbidity Index score simultaneously. This study suggested that offspring of long-lived siblings not only show lower CVD incidence but also a better survival following CVD diagnosis compared to matched population controls. The higher biological resilience appears to be a universal hallmark of longevity-enriched families, which makes them uniquely positioned for studying healthy aging and longevity mechanisms. Supplementary Information The online version contains supplementary material available at 10.1007/s10433-021-00641-7.
Collapse
Affiliation(s)
- Angéline Galvin
- Epidemiology, Biostatistics, and Biodemography, Department of Public Health, University of Southern Denmark, J.B. Winsløws Vej 9, 5 000 Odense C, Denmark
| | - Mary Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO USA
| | - Konstantin Arbeev
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC USA
| | - Allison L. Kuipers
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA USA
| | - Mary Wojczynski
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO USA
| | - Svetlana Ukrainsteva
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC USA
| | - Kaare Christensen
- Epidemiology, Biostatistics, and Biodemography, Department of Public Health, University of Southern Denmark, J.B. Winsløws Vej 9, 5 000 Odense C, Denmark
- Department of Public Health, The Danish Aging Research Center, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
23
|
Miljkovic I, Cvejkus R, An P, Thyagarajan B, Christensen K, Wojczynski M, Schupf N, Zmuda JM. Low Risk for Developing Diabetes Among the Offspring of Individuals With Exceptional Longevity and Their Spouses. FRONTIERS IN CLINICAL DIABETES AND HEALTHCARE 2022; 3:753986. [PMID: 36992755 PMCID: PMC10012150 DOI: 10.3389/fcdhc.2022.753986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 03/15/2022] [Indexed: 11/13/2022]
Abstract
Little is known about the risk of type 2 diabetes (T2D) among the offspring of individuals with exceptional longevity. We determined the incidence of and potential risk and protective factors for T2D among the offspring of probands and offspring’s spouses (mean age=60 years, range 32-88 years) in the Long Life Family Study (LLFS), a multicenter cohort study of 583 two-generation families with a clustering of healthy aging and exceptional longevity. Incident T2D was defined as fasting serum glucose ≥126 mg/dl, or HbA1c of ≥6.5%, or self-reported with doctor diagnosis of T2D, or the use of anti-diabetic medication during a mean follow-up 7.9 ± 1.1 years. Among offspring (n=1105) and spouses (n=328) aged 45-64 years without T2D at baseline visit, the annual incident rate of T2D was 3.6 and 3.0 per 1000 person-years, respectively, while among offspring (n=444) and spouses (n=153) aged 65+ years without T2D at baseline, the annual incident rate of T2D was 7.2 and 7.4 per 1000 person-years, respectively. By comparison, the annual incident rate of T2D per 1000 person-years in the U.S. general population was 9.9 among those aged 45-64, and 8.8 among those aged 65+ years (2018 National Health Interview Survey). Baseline BMI, waist circumference, and fasting serum triglycerides were positively associated with incident T2D, whereas fasting serum HDL-C, adiponectin, and sex hormone binding globulin were protective against incident T2D among the offspring (all P<0.05). Similar associations were observed among their spouses (all P<0.05, except sex hormone binding globulin). In addition, we observed that among spouses, but not offspring, fasting serum interleukin 6 and insulin-like growth factor 1 were positively associated with incident T2D (P<0.05 for both). Our study suggests that both offspring of long-living individuals and their spouses, especially middle-aged, share a similar low risk for developing T2D as compared with the general population. Our findings also raise the possibility that distinct biological risk and protective factors may contribute to T2D risk among offspring of long-lived individuals when compared with their spouses. Future studies are needed to identify the mechanisms underlying low T2D risk among the offspring of individuals with exceptional longevity, and also among their spouses.
Collapse
Affiliation(s)
- Iva Miljkovic
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
- *Correspondence: Iva Miljkovic,
| | - Ryan Cvejkus
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ping An
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, United States
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Kaare Christensen
- Department of Epidemiology, Biostatistics and Biodemography, Danish Aging Research Center, University of Southern Denmark, Odense, Denmark
| | - Mary Wojczynski
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, United States
| | - Nicole Schupf
- Taub Institute, Columbia University, New York, NY, United States
| | - Joseph M. Zmuda
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
24
|
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
|
25
|
The 90 plus: longevity and COVID-19 survival. Mol Psychiatry 2022; 27:1936-1944. [PMID: 35136227 DOI: 10.1038/s41380-022-01461-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/11/2022] [Accepted: 01/20/2022] [Indexed: 11/08/2022]
Abstract
The world population is getting older and studies aiming to enhance our comprehension of the underlying mechanisms responsible for health span are of utmost interest for longevity and as a measure for health care. In this review, we summarized previous genetic association studies (GWAS) and next-generation sequencing (NGS) of elderly cohorts. We also present the updated hypothesis for the aging process, together with the factors associated with healthy aging. We discuss the relevance of studying older individuals and build databanks to characterize the presence and resistance against late-onset disorders. The identification of about 2 million novel variants in our cohort of more than 1000 elderly Brazilians illustrates the importance of studying highly admixed populations of non-European ancestry. Finally, the ascertainment of nonagenarians and particularly of centenarians who were recovered from COVID-19 or remained asymptomatic opens new avenues of research aiming to enhance our comprehension of biological mechanisms associated with resistance against pathogens.
Collapse
|
26
|
Du M, Andersen SL, Cosentino S, Boudreau RM, Perls TT, Sebastiani P. Digitally generated Trail Making Test data: Analysis using hidden Markov modeling. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2022; 14:e12292. [PMID: 35280964 PMCID: PMC8902814 DOI: 10.1002/dad2.12292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 01/04/2022] [Accepted: 01/16/2022] [Indexed: 06/14/2023]
Abstract
The Trail Making Test (TMT) is a neuropsychological test used to assess cognitive dysfunction. The TMT consists of two parts: TMT-A requires connecting numbers 1 to 25 sequentially; TMT-B requires connecting numbers 1 to 12 and letters A to L sequentially, alternating between numbers and letters. We propose using a digitally recorded version of TMT to capture cognitive or physical functions underlying test performance. We analyzed digital versions of TMT-A and -B to derive time metrics and used Bayesian hidden Markov models to extract additional metrics. We correlated these derived metrics with cognitive and physical function scores using regression. On both TMT-A and -B, digital metrics associated with graphomotor processing test scores and gait speed. Digital metrics on TMT-B were additionally associated with episodic memory test scores and grip strength. These metrics provide additional information of cognitive state and can differentiate cognitive and physical factors affecting test performance.
Collapse
Affiliation(s)
- Mengtian Du
- Department of BiostatisticsBoston UniversityBostonMassachusettsUSA
- Analysis Group111 Huntington Ave. 14th floorBostonMA02119USA
| | - Stacy L. Andersen
- Department of MedicineBoston University School of MedicineBostonMassachusettsUSA
| | - Stephanie Cosentino
- Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
- Gertrude H. Sergievsky CenterColumbia UniversityNew YorkNew YorkUSA
| | - Robert M. Boudreau
- Department of EpidemiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Thomas T. Perls
- Department of MedicineBoston University School of MedicineBostonMassachusettsUSA
| | - Paola Sebastiani
- Institute for Clinical Research and Health Policy StudiesTufts Medical CenterBostonMassachusettsUSA
| |
Collapse
|
27
|
Renner SW, Qiao Y(S, Gmelin T, Santanasto AJ, Boudreau RM, Walston JD, Perls TT, Christensen K, Newman AB, Glynn NW. Association of fatigue, inflammation, and physical activity on gait speed: the Long Life Family Study. Aging Clin Exp Res 2022; 34:367-374. [PMID: 34196949 PMCID: PMC8864668 DOI: 10.1007/s40520-021-01923-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 06/20/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND Fatigue, inflammation, and physical activity (PA) are all independently associated with gait speed, but their directionality is not fully elucidated. AIMS Evaluate the bidirectional associations amongst fatigue, inflammation, and PA on gait speed. METHODS This cross sectional study included probands (n = 1280, aged 49-105) and offspring (n = 2772, aged 24-88) in the Long Life Family Study. We assessed gait speed, fatigue with the question "I could not get going", inflammation using fasting interleukin-6 (IL-6) and high sensitivity C-reactive protein (CRP), and self-reported PA as walking frequency in the past two weeks. The two generations were examined separately using linear mixed modeling. RESULTS Lower fatigue, lower IL-6, and greater PA were all associated with faster gait speed in both generations (all p < 0.05); lower CRP was only associated with faster gait speed in the offspring. PA explained the association of fatigue and gait speed via a 16.1% (95% CI 9.7%, 26.7%) attenuation of the direct associations for the probands and 9.9% (95% CI 6.3%, 18.8%) in the offspring. In addition, IL-6 explained more of the association of fatigue and gait speed than the association between PA and gait speed, via a 14.9% (95% CI 9.2%, 23.4%) attenuation of the direct association in the offspring only. DISCUSSION Results revealed a potential directionality from fatigue to IL-6 to PA that may lead to faster gait speed. Future work should examine these relationships longitudinally to establish temporality and causality. CONCLUSIONS Our findings support a signal that lowering fatigue and inflammation and increasing physical activity may delay functional decline.
Collapse
Affiliation(s)
- Sharon W. Renner
- University of Pittsburgh, Graduate School of Public Health, Department of Epidemiology, Pittsburgh, Pennsylvania
| | - Yujia (Susanna) Qiao
- University of Pittsburgh, Graduate School of Public Health, Department of Epidemiology, Pittsburgh, Pennsylvania
| | - Theresa Gmelin
- University of Pittsburgh, Graduate School of Public Health, Department of Epidemiology, Pittsburgh, Pennsylvania
| | - Adam J. Santanasto
- University of Pittsburgh, Graduate School of Public Health, Department of Epidemiology, Pittsburgh, Pennsylvania
| | - Robert M. Boudreau
- University of Pittsburgh, Graduate School of Public Health, Department of Epidemiology, Pittsburgh, Pennsylvania
| | - Jeremy D. Walston
- Johns Hopkins School of Medicine, Division of Geriatrics and Gerontology, Baltimore, Maryland
| | - Thomas T. Perls
- Boston University School of Medicine, Department of Medicine, Geriatrics Section, Boston, Massachusetts
| | - Kaare Christensen
- University of Southern Denmark, Department of Public Health, Unit of Epidemiology, Biostatistics and Biodemography, Odense, Denmark
| | - Anne B. Newman
- University of Pittsburgh, Graduate School of Public Health, Department of Epidemiology, Pittsburgh, Pennsylvania
| | - Nancy W. Glynn
- University of Pittsburgh, Graduate School of Public Health, Department of Epidemiology, Pittsburgh, Pennsylvania
| | | |
Collapse
|
28
|
Wojczynski MK, Lin SJ, Sebastiani P, Perls TT, Lee J, Kulminski A, Newman A, Zmuda JM, Christensen K, Province MA. NIA Long Life Family Study: Objectives, Design, and Heritability of Cross Sectional and Longitudinal Phenotypes. J Gerontol A Biol Sci Med Sci 2021; 77:717-727. [PMID: 34739053 PMCID: PMC8974329 DOI: 10.1093/gerona/glab333] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Indexed: 12/02/2022] Open
Abstract
The NIA Long Life Family Study (LLFS) is a longitudinal, multicenter, multinational, population-based multigenerational family study of the genetic and nongenetic determinants of exceptional longevity and healthy aging. The Visit 1 in-person evaluation (2006–2009) recruited 4 953 individuals from 539 two-generation families, selected from the upper 1% tail of the Family Longevity Selection Score (FLoSS, which quantifies the degree of familial clustering of longevity). Demographic, anthropometric, cognitive, activities of daily living, ankle-brachial index, blood pressure, physical performance, and pulmonary function, along with serum, plasma, lymphocytes, red cells, and DNA, were collected. A Genome Wide Association Scan (GWAS) (Ilumina Omni 2.5M chip) followed by imputation was conducted. Visit 2 (2014–2017) repeated all Visit 1 protocols and added carotid ultrasonography of atherosclerotic plaque and wall thickness, additional cognitive testing, and perceived fatigability. On average, LLFS families show healthier aging profiles than reference populations, such as the Framingham Heart Study, at all age/sex groups, for many critical healthy aging phenotypes. However, participants are not uniformly protected. There is considerable heterogeneity among the pedigrees, with some showing exceptional cognition, others showing exceptional grip strength, others exceptional pulmonary function, etc. with little overlap in these families. There is strong heritability for key healthy aging phenotypes, both cross-sectionally and longitudinally, suggesting that at least some of this protection may be genetic. Little of the variance in these heritable phenotypes is explained by the common genome (GWAS + Imputation), which may indicate that rare protective variants for specific phenotypes may be running in selected families.
Collapse
Affiliation(s)
- Mary K Wojczynski
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
| | - Shiow Jiuan Lin
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
| | - Paola Sebastiani
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Thomas T Perls
- Department of Medicine, Geriatrics Section, Boston Medical Center, Boston University School of Medicine, MA, USA
| | - Joseph Lee
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Alexander Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, USA
| | - Anne Newman
- Departments of Epidemiology and Human Genetics, Graduate School of Public Health, University of Pittsburgh, PA, USA
| | - Joe M Zmuda
- Departments of Epidemiology and Human Genetics, Graduate School of Public Health, University of Pittsburgh, PA, USA
| | - Kaare Christensen
- Unit of Epidemiology, Biostatistics and Biodemography, Department of Public Health, Southern Denmark University, Odense, Denmark
| | - Michael A Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
29
|
Du M, Andersen SL, Schupf N, Feitosa MF, Barker MS, Perls TT, Sebastiani P. Association Between APOE Alleles and Change of Neuropsychological Tests in the Long Life Family Study. J Alzheimers Dis 2021; 79:117-125. [PMID: 33216038 DOI: 10.3233/jad-201113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND The Long Life Family Study (LLFS) is a family based, prospective study of healthy aging and familial longevity. The study includes two assessments of cognitive function that were administered approximately 8 years apart. OBJECTIVE To test whether APOE genotype is associated with change of cognitive function in older adults. METHODS We used Bayesian hierarchical models to test the association between APOE alleles and change of cognitive function. Six longitudinally collected neuropsychological test scores were modelled as a function of age at enrollment, follow-up time, gender, education, field center, birth cohort indicator (≤1935, or >1935), and the number of copies of ɛ2 or ɛ4 alleles. RESULTS Out of 4,587 eligible participants, 2,064 were male (45.0%), and age at enrollment ranged from 25 to 110 years, with mean of 70.85 years (SD: 15.75). We detected a significant cross-sectional effect of the APOEɛ4 allele on Logical Memory. Participants carrying at least one copy of the ɛ4 allele had lower scores in both immediate (-0.31 points, 95% CI: -0.57, -0.05) and delayed (-0.37 points, 95% CI: -0.64, -0.10) recall comparing to non-ɛ4 allele carriers. We did not detect any significant longitudinal effect of the ɛ4 allele. There was no cross-sectional or longitudinal effect of the ɛ2 allele. CONCLUSION The APOEɛ4 allele was identified as a risk factor for poorer episodic memory in older adults, while the APOEɛ2 allele was not significantly associated with any of the cognitive test scores.
Collapse
Affiliation(s)
- Mengtian Du
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Stacy L Andersen
- Geriatrics Section, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Nicole Schupf
- Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, and the Gertrude H. Sergievsky Center, Columbia University Medical Center, New York, NY, USA.,Department of Epidemiology, Columbia University Mailman School of Public Health, Sergievsky Center, New York, NY, USA
| | - Mary F Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Megan S Barker
- Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, and the Gertrude H. Sergievsky Center, Columbia University Medical Center, New York, NY, USA
| | - Thomas T Perls
- Geriatrics Section, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Paola Sebastiani
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA, USA
| |
Collapse
|
30
|
Qiao Y(S, Gmelin T, Renner SW, Boudreau RM, Martin S, Wojczynski MK, Christensen K, Andersen SL, Cosentino S, Santanasto AJ, Glynn NW. Evaluation of the Bidirectional Relations of Perceived Physical Fatigability and Physical Activity on Slower Gait Speed. J Gerontol A Biol Sci Med Sci 2021; 76:e237-e244. [PMID: 33170216 PMCID: PMC8436994 DOI: 10.1093/gerona/glaa281] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Lower physical activity levels and greater fatigability contribute independently to slower gait speed in older adults. To fully understand the bidirectional relations between physical activity and fatigability, and to inform potential intervention strategies, we examined whether physical activity or fatigability explains more of the other factor's association on slower gait speed. METHODS Two generations (probands and offspring) of older adults (N = 2079, mean age 73.0 ± 10.0 years, 54.2% women, 99.7% White) enrolled in the Long Life Family Study were assessed at Visit 2 (2014-2017). Self-reported physical activity was measured with the Framingham Physical Activity Index and perceived physical fatigability using the Pittsburgh Fatigability Scale. Statistical mediation analyses were conducted separately by generation with linear mixed-effect models accounting for family relatedness and adjusted for demographics, health conditions, and field center. RESULTS Greater perceived physical fatigability explained the association of lower physical activity on slower gait speed via a 22.5% attenuation of the direct association (95% confidence interval [CI]: 15.0%-35.2%) for the probands and 39.5% (95% CI: 22.8%-62.6%) for the offspring. Whereas lower physical activity explained the association of greater perceived fatigability on slower gait speed via a 22.5% attenuation of the direct association (95% CI: 13.4%-32.8%) for the probands and 6.7% (95% CI: 3.8%-15.4%) for the offspring. CONCLUSIONS Our findings suggest that the impact of greater perceived physical fatigability on the association between lower physical activity and slower gait speed differs between younger-old and middle-to-oldest-old adults, indicating perceived physical fatigability as a potential mediator in the disablement pathway.
Collapse
Affiliation(s)
| | - Theresa Gmelin
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sharon W Renner
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Robert M Boudreau
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sarah Martin
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mary K Wojczynski
- Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | - Kaare Christensen
- Institute of Public Health, University of Southern Denmark, Odense, Denmark
| | - Stacy L Andersen
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Stephanie Cosentino
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Medical Center, New York, New York
| | - Adam J Santanasto
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Nancy W Glynn
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
31
|
Santanasto AJ, Wojczynski MK, Cvejkus RK, Lin S, Wang L, Thyagarajan B, Christensen K, Schupf N, Feitosa MF, An P, Zmuda JM. Identification of a Novel Locus for Gait Speed Decline With Aging: The Long Life Family Study. J Gerontol A Biol Sci Med Sci 2021; 76:e307-e313. [PMID: 34156441 PMCID: PMC8436996 DOI: 10.1093/gerona/glab177] [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: 12/11/2020] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Gait speed is a powerful indicator of health with aging. Potential genetic contributions to gait speed and its decline with aging are not well defined. We determined the heritability of and potential genetic regions underlying change in gait speed using longitudinal data from 2379 individuals belonging to 509 families in the Long Life Family Study (mean age 64 ± 12, range 30-110 years; 45% men). METHODS Gait speed was measured over 4 m at baseline and follow-up (7 ± 1 years). Quantitative trait linkage analyses were completed using pedigree-based maximum likelihood methods with logarithm of the odds (LOD) scores greater than 3.0, indicating genome-wide significance. We also performed linkage analysis in the top 10% of families contributing to LOD scores to allow for heterogeneity among families (HLOD). Data were adjusted for age, sex, height, and field center. RESULTS At baseline, 26.9% of individuals had "slow" gait speed less than 1.0 m/s (mean: 1.1 ± 0.2 m/s) and gait speed declined at a rate of -0.02 ± 0.03 m/s per year (p < .0001). Baseline and change in gait speed were significantly heritable (h2 = 0.24-0.32, p < .05). We did not find significant evidence for linkage for baseline gait speed; however, we identified a significant locus for change in gait speed on chromosome 16p (LOD = 4.2). A subset of 21 families contributed to this linkage peak (HLOD = 6.83). Association analyses on chromosome 16 showed that the strongest variant resides within the ADCY9 gene. CONCLUSION Further analysis of the chromosome 16 region, and ADCY9 gene, may yield new insight on the biology of mobility decline with aging.
Collapse
Affiliation(s)
- Adam J Santanasto
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pennsylvania, USA
| | - Mary K Wojczynski
- Department of Genetics, Division of Statistical Genomics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ryan K Cvejkus
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pennsylvania, USA
| | - Shiow Lin
- Department of Genetics, Division of Statistical Genomics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Lihua Wang
- Department of Genetics, Division of Statistical Genomics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Minnesota, Minneapolis, USA
| | - Kaare Christensen
- The Danish Aging Research Center, Epidemiology Unit, Institute of Public Health, University of Southern Denmark, Odense, Denmark
| | - Nicole Schupf
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, USA
| | - Mary F Feitosa
- Department of Genetics, Division of Statistical Genomics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ping An
- Department of Genetics, Division of Statistical Genomics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Joseph M Zmuda
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pennsylvania, USA
| |
Collapse
|
32
|
Yudkovicz JJ, Minster RL, Barinas-Mitchell E, Christensen K, Feitosa M, Barker MS, Newman AB, Kuipers AL. Pleiotropic effects between cardiovascular disease risk factors and measures of cognitive and physical function in long-lived adults. Sci Rep 2021; 11:17980. [PMID: 34504188 PMCID: PMC8429644 DOI: 10.1038/s41598-021-97298-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 08/17/2021] [Indexed: 11/16/2022] Open
Abstract
Cardiovacular disease (CVD) is the leading cause of death among older adults and is often accompanied by functional decline. It is unclear what is driving this co-occurrence, but it may be behavioral, environmental and/or genetic. We used a family-based study to estimate the phenotypic and shared genetic correlation between CVD risk factors and physical and cognitive functional measures. Participants (n = 1,881) were from the Long Life Family Study, which enrolled families based on their exceptional longevity (sample mean age = 69.4 years, 44% female). Cardiovascular disease risk factors included carotid vessel measures [intima-media thickness and inter-adventitial diameter], obesity [body mass index (BMI) and waist circumference], and hypertension [systolic and diastolic blood pressures]. Function was measured in the physical [gait speed, grip strength, chair stand] and cognitive [digital symbol substitution test, retained and working memory, semantic fluency, and trail making tests] domains. We used SOLAR to estimate the genetic, environmental, and phenotypic correlation between each pair adjusting for age, age2, sex, field center, smoking, height, and weight. There were significant phenotypic correlations (range |0.05–0.22|) between CVD risk factors and physical and cognitive function (all P < 0.05). Most significant genetic correlations (range |0.21–0.62|) were between CVD risk factorsand cognitive function, although BMI and waist circumference had significant genetic correlation with gait speed and chair stand time (range |0.29–0.53|; all P < 0.05). These results suggest that CVD risk factors may share a common genetic-and thus, biologic-basis with both cognitive and physical function. This is particularly informative for research into the genetic determinants of chronic disease.
Collapse
Affiliation(s)
- Julia J Yudkovicz
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ryan L Minster
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Kaare Christensen
- Department of Epidemiology, Biostatistics and Biodemography, Danish Aging Research Center, University of Southern Denmark, Odense C, Denmark
| | - Mary Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Megan S Barker
- Department of Neurology, Columbia University, New York, NY, USA
| | - Anne B Newman
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Allison L Kuipers
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
33
|
Libon DJ, Baliga G, Swenson R, Au R. Digital Neuropsychological Assessment: New Technology for Measuring Subtle Neuropsychological Behavior. J Alzheimers Dis 2021; 82:1-4. [PMID: 34219670 DOI: 10.3233/jad-210513] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Technology has transformed the science and practice of medicine. In this special mini-forum, data using digital neuropsychological technology are reported. All of these papers demonstrate how coupling digital technology with standard paper and pencil neuropsychological tests are able to extract behavior not otherwise obtainable. As digital assessment methods mature, early identification of persons with emergent neurodegenerative and other neurological illness may be possible.
Collapse
Affiliation(s)
- David J Libon
- Department Geriatrics, Gerontology, and Psychology, New Jersey Institute for Successful Aging, School of Osteopathic Medicine, Rowan University, Glassboro, NJ, USA
| | - Ganesh Baliga
- Department of Computer Science, Rowan University, Glassboro, NJ, USA
| | - Rod Swenson
- Department Psychiatry and Behavioral Science, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Rhoda Au
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA.,Framingham Heart Study, Boston University School of Medicine, Boston, MA, USA.,Department of Neurology, Boston University School of Medicine, Boston, MA, USA.,Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| |
Collapse
|
34
|
Andersen SL, Sweigart B, Glynn NW, Wojczynski MK, Thyagarajan B, Mengel-From J, Thielke S, Perls TT, Libon DJ, Au R, Cosentino S, Sebastiani P. Digital Technology Differentiates Graphomotor and Information Processing Speed Patterns of Behavior. J Alzheimers Dis 2021; 82:17-32. [PMID: 34219735 DOI: 10.3233/jad-201119] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Coupling digital technology with traditional neuropsychological test performance allows collection of high-precision metrics that can clarify and/or define underlying constructs related to brain and cognition. OBJECTIVE To identify graphomotor and information processing trajectories using a digitally administered version of the Digit Symbol Substitution Test (DSST). METHODS A subset of Long Life Family Study participants (n = 1,594) completed the DSST. Total time to draw each symbol was divided into 'writing' and non-writing or 'thinking' time. Bayesian clustering grouped participants by change in median time over intervals of eight consecutively drawn symbols across the 90 s test. Clusters were characterized based on sociodemographic characteristics, health and physical function data, APOE genotype, and neuropsychological test scores. RESULTS Clustering revealed four 'thinking' time trajectories, with two clusters showing significant changes within the test. Participants in these clusters obtained lower episodic memory scores but were similar in other health and functional characteristics. Clustering of 'writing' time also revealed four performance trajectories where one cluster of participants showed progressively slower writing time. These participants had weaker grip strength, slower gait speed, and greater perceived physical fatigability, but no differences in cognitive test scores. CONCLUSION Digital data identified previously unrecognized patterns of 'writing' and 'thinking' time that cannot be detected without digital technology. These patterns of performance were differentially associated with measures of cognitive and physical function and may constitute specific neurocognitive biomarkers signaling the presence of subtle to mild dysfunction. Such information could inform the selection and timing of in-depth neuropsychological assessments and help target interventions.
Collapse
Affiliation(s)
- Stacy L Andersen
- Geriatrics Section, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Benjamin Sweigart
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Nancy W Glynn
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mary K Wojczynski
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota School of Medicine, Minneapolis, MN, USA
| | - Jonas Mengel-From
- Institute of Public Health, Epidemiology, Biostatistics and Biodemography Unit, University of Southern Denmark, Odense, Denmark
| | - Stephen Thielke
- Geriatric Research, Education, and Clinical Center, Puget Sound VA Medical Center, Seattle, WA, USA
| | - Thomas T Perls
- Geriatrics Section, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - David J Libon
- New Jersey Institute for Successful Aging, School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
| | - Rhoda Au
- Department of Anatomy and Neurobiology and Neurology, Boston University School of Medicine, Boston, MA, USA.,Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Stephanie Cosentino
- Cognitive Neuroscience Division of the Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA.,Gertrude H. Sergievsky Center, Columbia University, New York, NY, USA
| | - Paola Sebastiani
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA, USA
| | | |
Collapse
|
35
|
Santanasto AJ, Cvejkus RK, Wojczynski MK, Marron MM, Schupf N, Christensen K, Thyagarajan B, Zmuda JM. Circulating Procollagen Type III N-Terminal Peptide and Physical Function in Adults from the Long Life Family Study. J Gerontol A Biol Sci Med Sci 2021; 76:1273-1279. [PMID: 32794566 PMCID: PMC8355442 DOI: 10.1093/gerona/glaa197] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Circulating levels of procollagen type III N-terminal peptide (P3NP) may reflect increased fibrosis of skeletal muscle and other tissues with aging. Herein, we tested if P3NP was associated with baseline and 7-year change in physical function. METHOD Participants (n = 400) were from the Long Life Family Study, a study of exceptional familial longevity. Plasma P3NP concentration was measured using a sandwich enzyme-linked immunosorbent assay (inter-assay coefficient of variation <5.5%). At baseline and 7-year follow-up visits, physical function was measured using the Short Physical Performance Battery (SPPB score 0-12), which consists of gait speed, balance, and chair-rise tests. Grip strength was measured using a handheld dynamometer. The association between log-transformed P3NP and physical function was examined using generalized estimating equations adjusted for familial relatedness, age, sex, height, weight, lifestyle characteristics, liver function, kidney function, lung function, and chronic disease prevalence. RESULTS Participants were aged 73.1 ± 15.2 years (range: 39-104), 54% female, had body mass index of 26.6 ± 4.3 kg/m2, and gait speeds of 1.0 ± 0.3 m/s. One standard deviation higher log-transformed P3NP was related to worse baseline SPPB score (β = -0.9points), gait speed (β = -0.05m/s), chair-rises per-second (β = -0.46chair-rises/10 seconds), and grip strength (β = -2.0kg; all p < .001). Higher P3NP was also associated with greater declines in gait speed (β = -1.41, p < .001) and transitioning to being unable to perform chair-rises (β = 0.41, p < .001) after 7 years. CONCLUSION Plasma P3NP may be a strong, novel biomarker of current and future physical function. Future research is needed to extend our findings to other cohorts and determine mechanisms underlying these associations.
Collapse
Affiliation(s)
- Adam J Santanasto
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pennsylvania
| | - Ryan K Cvejkus
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pennsylvania
| | - Mary K Wojczynski
- Department of Genetics, Division of Statistical Genomics, Washington University School of Medicine in St. Louis, Missouri
| | - Megan M Marron
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pennsylvania
| | - Nicole Schupf
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York City, New York
| | - Kaare Christensen
- The Danish Aging Research Center, Epidemiology Unit, Institute of Public Health, University of Southern Denmark, Odense
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Minnesota, Minneapolis
| | - Joseph M Zmuda
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pennsylvania
| |
Collapse
|
36
|
Andersen SL, Du M, Cosentino S, Schupf N, Rosso AL, Perls TT, Sebastiani P. Slower Decline in Processing Speed Is Associated with Familial Longevity. Gerontology 2021; 68:17-29. [PMID: 33946077 PMCID: PMC9093735 DOI: 10.1159/000514950] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 02/03/2021] [Indexed: 01/03/2023] Open
Abstract
INTRODUCTION Cross-sectional analyses have associated familial longevity with better cognitive function and lower risk of cognitive impairment in comparison with individuals without familial longevity. The extent to which long-lived families also demonstrate slower rates of cognitive aging (i.e., change in cognition over time) is unknown. This study examined longitudinally collected data among 2 generations of the Long Life Family Study (LLFS) to compare rates of cognitive change across relatives and spouse controls. METHODS We analyzed change in 6 neuropsychological test scores collected approximately 8 years apart among LLFS family members (n = 3,972) versus spouse controls (n = 1,092) using a Bayesian hierarchical model that included age, years of follow-up, sex, education, generation, and field center and all possible pairwise interactions. RESULTS At a mean age of 88 years at enrollment in the older generation and 60 years in the younger generation, LLFS family members performed better than their spouses on the Digit Symbol Substitution Test (DSST) and the Logical Memory test. At follow-up, family members in the younger generation also showed slower decline than spouses on the DSST, whereas rates of change of Digit Span, fluency, and memory were similar between the 2 groups. DISCUSSION/CONCLUSION Individuals in families with longevity appear to have better cognitive performance than their spouses for cognitive processes including psychomotor processing, episodic memory, and retrieval. Additionally, they demonstrate longer cognitive health spans with a slower decline on a multifactorial test of processing speed, a task requiring the integration of processes including organized visual search, working and incidental memory, and graphomotor ability. Long-lived families may be a valuable cohort for studying resilience to cognitive aging.
Collapse
Affiliation(s)
| | - Mengtian Du
- Department of Biostatistics, Boston University School of Public Health
| | - Stephanie Cosentino
- Department of Neurology and Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University
- Gertrude H. Sergievsky Center, Columbia University
| | - Nicole Schupf
- Department of Neurology and Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University
- Gertrude H. Sergievsky Center, Columbia University
| | | | - Thomas T. Perls
- Department of Medicine, Boston University School of Medicine
| | - Paola Sebastiani
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center
| |
Collapse
|
37
|
Feitosa MF, Kuipers AL, Wojczynski MK, Wang L, Barinas-Mitchell E, Kulminski AM, Thyagarajan B, Lee JH, Perls T, Christensen K, Newman AB, Zmuda JM, Province MA. Heterogeneity of the Predictive Polygenic Risk Scores for Coronary Heart Disease Age-at-Onset in Three Different Coronary Heart Disease Family-Based Ascertainments. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2021; 14:e003201. [PMID: 33844929 DOI: 10.1161/circgen.120.003201] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Polygenic risk scores (PRS) for coronary heart disease (CHD) may contribute to assess the overall risk of CHD. We evaluated how PRS may influence CHD risk when the distribution of age-at-onset, sex, and family health history differ significantly. METHODS Our study included 3 family-based ascertainments: LLFS (Long Life Family Study, NIndividuals=4572), which represents a low CHD risk, and Family Heart Study, which consists of randomly selected families (FamHS-random, NIndividuals=1806), and high CHD risk families (FamHS-high risk, NIndividuals=2301). We examined the effects of PRS, sex, family ascertainment, PRS interaction with sex (PRS*sex) and with family ascertainment (PRS*LLFS and PRS*FamHS-high risk) on CHD, corrected for traditional cardiovascular risk factors using Cox proportional hazard regression models. RESULTS Healthy-aging LLFS presented ≈17 years delayed for CHD age-at-onset compared with FamHS-high risk (P<1.0×10-4). Sex-specific association (P<1.0×10-17) and PRS*sex (P=2.7×10-3) predicted prevalent CHD. CHD age-at-onset was associated with PRS (hazard ratio [HR], 1.57; P=1.3×10-5), LLFS (HR, 0.54; P=2.6×10-5), and FamHS-high risk (HR, 2.86; P=6.70x10-15) in men, and with PRS (HR, 1.76; P=7.70×10-3), FamHS-high risk (HR, 4.88; P=8.70×10-10), and PRS×FamHS-high risk (HR, 0.61; P=3.60×10-2) in women. In the PRS extreme quartile distributions, CHD age-at-onset was associated (P<0.05) with PRS, FamHS-high risk, and PRS interactions with both low and high CHD risk families for women. For men, the PRS quartile results remained similar to the whole distribution. CONCLUSIONS Differences in CHD family-based ascertainments show evidence of PRS interacting with sex to predict CHD risk. In women, CHD age-at-onset was associated with PRS, CHD family history, and interactions of PRS with family history. In men, PRS and CHD family history were the major effects on the CHD age-at-onset. Understanding the heterogeneity of risks associated with CHD end points at both the personal and familial levels may shed light on the underlying genetic effects influencing CHD and lead to more personalized risk prediction.
Collapse
Affiliation(s)
- Mary F Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO (M.F.F., M.K.W., L.W., M.A.P.)
| | - Allison L Kuipers
- Department of Epidemiology (A.L.K., E.B.-M., A.B.N., J.M.Z.), University of Pittsburgh, PA
| | - Mary K Wojczynski
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO (M.F.F., M.K.W., L.W., M.A.P.)
| | - Lihua Wang
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO (M.F.F., M.K.W., L.W., M.A.P.)
| | - Emma Barinas-Mitchell
- Department of Epidemiology (A.L.K., E.B.-M., A.B.N., J.M.Z.), University of Pittsburgh, PA
| | - Alexander M Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC (A.M.K.)
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis (B.T.)
| | - Joseph H Lee
- Sergievsky Center, Taub Institute, Department of Epidemiology and Department of Neurology, Columbia University, NY (J.H.L.)
| | - Thomas Perls
- Department of Medicine, Boston University School of Medicine, MA (T.P.)
| | - Kaare Christensen
- Danish Aging Research Center, University of Southern Denmark, Odense C (K.C.)
| | - Anne B Newman
- Department of Epidemiology (A.L.K., E.B.-M., A.B.N., J.M.Z.), University of Pittsburgh, PA
| | - Joseph M Zmuda
- Department of Epidemiology (A.L.K., E.B.-M., A.B.N., J.M.Z.), University of Pittsburgh, PA.,Department of Human Genetics (J.M.Z.), University of Pittsburgh, PA
| | - Michael A Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO (M.F.F., M.K.W., L.W., M.A.P.)
| |
Collapse
|
38
|
Yang Y, Zmuda JM, Wojczynski MK, Thyagarajan B, Christensen K, Cvejkus RK, Kuipers AL. Genetic association analysis of the cardiovascular biomarker: N-terminal fragment of pro-B-type natriuretic peptide (NT-proBNP). PLoS One 2021; 16:e0248726. [PMID: 33720941 PMCID: PMC7959346 DOI: 10.1371/journal.pone.0248726] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 03/03/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND NT-proBNP is a biomarker of cardiovascular disease (CVD). Little is known about the heritability and genetic variants associated with NT-proBNP. Therefore, we estimated the heritability of and examined genetic associations of SNPs in the BNP gene region with circulating NT-proBNP and prevalent CVD in 4,331 participants from the Long Life Family Study (LLFS). METHODS AND RESULTS Genotypes of 10 SNPs from the NPPB and NPPA regions that encode BNP and A-type natriuretic peptide, respectively, were tested for association with NT-proBNP and prevalent cardiovascular disease and risk factors. We performed analyses using the Sequential Oligogenic Linkage Analysis (SOLAR) program to account for family relatedness, and adjusted all models for age, sex, and field center. The mean age of the LLFS was 69.8 years (range 24-110) with 55.4% females. NT-proBNP was significantly heritable (h2 = 0.21; P = 4x10-14), and the minor alleles of rs632793 (p<0.001) and rs41300100 (p = 0.05) were independently associated with higher serum NT-proBNP levels. Additionally, the minor allele of rs632793 was significantly and consistently associated with lower prevalent CVD, including blood pressures, independent of NT-proBNP level (all P<0.05). Results for prevalent CVD, but not NT-proBNP levels, showed significant interaction by familial generation. CONCLUSION In this family-based study of subjects with exceptional longevity, we identified several allelic variants in the BNP gene region associated with NT-pro-BNP levels and prevalent CVD.
Collapse
Affiliation(s)
- Yuan Yang
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Joseph M. Zmuda
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Mary K. Wojczynski
- Department of Genetics, Washington University in St Louis, St. Louis, MO, United States of America
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States of America
| | - Kaare Christensen
- Department of Epidemiology, Biostatistics and Biodemography, Danish Aging Research Center, University of Southern Denmark, Odense C, Denmark
| | - Ryan K. Cvejkus
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Allison L. Kuipers
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, United States of America
- * E-mail:
| |
Collapse
|
39
|
Rodríguez-Girondo M, van den Berg N, Hof MH, Beekman M, Slagboom E. Improved selection of participants in genetic longevity studies: family scores revisited. BMC Med Res Methodol 2021; 21:7. [PMID: 33407157 PMCID: PMC7789146 DOI: 10.1186/s12874-020-01193-7] [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: 06/03/2020] [Accepted: 12/14/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Although human longevity tends to cluster within families, genetic studies on longevity have had limited success in identifying longevity loci. One of the main causes of this limited success is the selection of participants. Studies generally include sporadically long-lived individuals, i.e. individuals with the longevity phenotype but without a genetic predisposition for longevity. The inclusion of these individuals causes phenotype heterogeneity which results in power reduction and bias. A way to avoid sporadically long-lived individuals and reduce sample heterogeneity is to include family history of longevity as selection criterion using a longevity family score. A main challenge when developing family scores are the large differences in family size, because of real differences in sibship sizes or because of missing data. METHODS We discussed the statistical properties of two existing longevity family scores: the Family Longevity Selection Score (FLoSS) and the Longevity Relatives Count (LRC) score and we evaluated their performance dealing with differential family size. We proposed a new longevity family score, the mLRC score, an extension of the LRC based on random effects modeling, which is robust for family size and missing values. The performance of the new mLRC as selection tool was evaluated in an intensive simulation study and illustrated in a large real dataset, the Historical Sample of the Netherlands (HSN). RESULTS Empirical scores such as the FLOSS and LRC cannot properly deal with differential family size and missing data. Our simulation study showed that mLRC is not affected by family size and provides more accurate selections of long-lived families. The analysis of 1105 sibships of the Historical Sample of the Netherlands showed that the selection of long-lived individuals based on the mLRC score predicts excess survival in the validation set better than the selection based on the LRC score . CONCLUSIONS Model-based score systems such as the mLRC score help to reduce heterogeneity in the selection of long-lived families. The power of future studies into the genetics of longevity can likely be improved and their bias reduced, by selecting long-lived cases using the mLRC.
Collapse
Affiliation(s)
- Mar Rodríguez-Girondo
- Department of Biomedical Data Sciences, section of Medical Statistics, Leiden University Medical Center, Albinusdreef 2, 2333, ZA, Leiden, the Netherlands.
| | - Niels van den Berg
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, Albinusdreef 2, 2333, ZA, Leiden, the Netherlands
| | - Michel H Hof
- Department of Clinical Epidemiology, Biostatistics, and Bioinformatics, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105, AZ, Amsterdam, the Netherlands
| | - Marian Beekman
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, Albinusdreef 2, 2333, ZA, Leiden, the Netherlands
| | - Eline Slagboom
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, Albinusdreef 2, 2333, ZA, Leiden, the Netherlands
| |
Collapse
|
40
|
Sweigart B, Andersen SL, Gurinovich A, Cosentino S, Schupf N, Perls TT, Sebastiani P. APOE E2/E2 Is Associated with Slower Rate of Cognitive Decline with Age. J Alzheimers Dis 2021; 83:853-860. [PMID: 34366332 PMCID: PMC8896199 DOI: 10.3233/jad-201205] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND The E4 allele of the APOE gene is known to be associated with cognitive impairment. However, a limited number of studies have examined the association between the E2 allele and longitudinal changes of cognitive function. OBJECTIVE To determine whether rates of cognitive change differ in carriers of the APOE E2 allele compared to other genotypes. METHODS We conducted a secondary analysis of data from two ongoing longitudinal cohort studies, the Long Life Family Study (LLFS) and New England Centenarian Study (NECS). We included participants who had APOE genotyping data, data from longitudinal administrations of the Telephone Interview for Cognitive Status (TICS), and age, sex, and education available. We assessed whether cognitive change as measured by rate of decline in TICS score differed among people with different APOE genotypes. We used a hierarchical mixed effect model with APOE genotypes, their interactions with age, and potential confounders. RESULTS After adjusting for sex and education, in carriers of the common E3/E3 genotype, TICS score decreased by 0.15 points per year of age. In those with the E2/E2 genotype, TICS score decreased by 0.05 points per year of age, a significantly slower rate of decline (p = 0.017). We observed no protective effect of the E2/E3 genotype on cognitive decline. CONCLUSION These results suggest a protective effect of the E2/E2 genotype on a measure of global cognitive function.
Collapse
Affiliation(s)
- Benjamin Sweigart
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Stacy L. Andersen
- Geriatrics Section, Department of Medicine, Boston University School of Medicine, Boston, MA
| | - Anastasia Gurinovich
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA
| | - Stephanie Cosentino
- Cognitive Neuroscience Division of the Department of Neurology and Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY
- Gertrude H. Sergievsky Center, Columbia University, New York, NY
| | - Nicole Schupf
- Cognitive Neuroscience Division of the Department of Neurology and Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY
- Gertrude H. Sergievsky Center, Columbia University, New York, NY
| | - Thomas T. Perls
- Geriatrics Section, Department of Medicine, Boston University School of Medicine, Boston, MA
| | - Paola Sebastiani
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA
| |
Collapse
|
41
|
Leukocyte Telomere Length Is Unrelated to Cognitive Performance Among Non-Demented and Demented Persons: An Examination of Long Life Family Study Participants. J Int Neuropsychol Soc 2020; 26:906-917. [PMID: 32342830 PMCID: PMC7983066 DOI: 10.1017/s1355617720000363] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVE Leukocyte telomere length (LTL) is a widely hypothesized biomarker of biological aging. Persons with shorter LTL may have a greater likelihood of developing dementia. We investigate whether LTL is associated with cognitive function, differently for individuals without cognitive impairment versus individuals with dementia or incipient dementia. METHOD Enrolled subjects belong to the Long Life Family Study (LLFS), a multi-generational cohort study, where enrollment was predicated upon exceptional family longevity. Included subjects had valid cognitive and telomere data at baseline. Exclusion criteria were age ≤ 60 years, outlying LTL, and missing sociodemographic/clinical information. Analyses were performed using linear regression with generalized estimating equations, adjusting for sex, age, education, country, generation, and lymphocyte percentage. RESULTS Older age and male gender were associated with shorter LTL, and LTL was significantly longer in family members than spouse controls (p < 0.005). LTL was not associated with working or episodic memory, semantic processing, and information processing speed for 1613 cognitively unimpaired individuals as well as 597 individuals with dementia or incipient dementia (p < 0.005), who scored significantly lower on all cognitive domains (p < 0.005). CONCLUSIONS Within this unique LLFS cohort, a group of families assembled on the basis of exceptional survival, LTL is unrelated to cognitive ability for individuals with and without cognitive impairment. LTL does not change in the context of degenerative disease for these individuals who are biologically younger than the general population.
Collapse
|
42
|
Christensen K, Wojczynski MK, Pedersen JK, Larsen LA, Kløjgaard S, Skytthe A, McGue M, Vaupel JW, Province MA. Mechanisms underlying familial aggregation of exceptional health and survival: A three-generation cohort study. Aging Cell 2020; 19:e13228. [PMID: 32886847 PMCID: PMC7576291 DOI: 10.1111/acel.13228] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 07/29/2020] [Accepted: 08/11/2020] [Indexed: 01/26/2023] Open
Abstract
The familial resemblance in length of adult life is very modest. Studies of parent-offspring and twins suggest that exceptional health and survival have a stronger genetic component than lifespan generally. To shed light on the underlying mechanisms, we collected information on Danish long-lived siblings (born 1886-1938) from 659 families, their 5379 offspring (born 1917-1982), and 10,398 grandchildren (born 1950-2010) and matched background population controls through the Danish 1916 Census, the Civil Registration System, the National Patient Register, and the Register of Causes of Death. Comparison with the background, population revealed consistently lower occurrence of almost all disease groups and causes of death in the offspring and the grandchildren. The expected incidence of hospitalization for mental and behavioral disorders was reduced by half in the offspring (hazard ratio 0.53, 95% confidence interval 0.45-0.62) and by one-third in the grandchildren (0.69, 0.61-0.78), while the numbers for tobacco-related cancer were 0.60 (0.51-0.70) and 0.71 (0.48-1.05), respectively. Within-family analyses showed a general, as opposed to specific, lowering of disease risk. Early parenthood and divorce were markedly less frequent in the longevity-enriched families, while economic and educational differences were small to moderate. The longevity-enriched families in this study have a general health advantage spanning three generations. The particularly low occurrence of mental and behavioral disorders and tobacco-related cancers together with indicators of family stability and only modest socioeconomic advantage implicate behavior as a key mechanism underlying familial aggregation of exceptional health and survival.
Collapse
Affiliation(s)
- Kaare Christensen
- Department of Public HealthDanish Aging Research CenterUniversity of Southern DenmarkOdenseDenmark
| | - Mary K. Wojczynski
- Department of GeneticsWashington University School of MedicineSt. LouisMOUSA
| | - Jacob K. Pedersen
- Department of Public HealthDanish Aging Research CenterUniversity of Southern DenmarkOdenseDenmark
| | - Lisbeth A. Larsen
- Department of Public HealthDanish Aging Research CenterUniversity of Southern DenmarkOdenseDenmark
| | - Susanne Kløjgaard
- Department of Public HealthDanish Aging Research CenterUniversity of Southern DenmarkOdenseDenmark
| | - Axel Skytthe
- Department of Public HealthDanish Aging Research CenterUniversity of Southern DenmarkOdenseDenmark
| | - Matt McGue
- Department of Public HealthDanish Aging Research CenterUniversity of Southern DenmarkOdenseDenmark
- Department of PsychologyUniversity of MinnesotaMinneapolisMNUSA
| | - James W. Vaupel
- Center on Population DynamicsUniversity of Southern DenmarkOdenseDenmark
| | - Michael A. Province
- Department of GeneticsWashington University School of MedicineSt. LouisMOUSA
| |
Collapse
|
43
|
Sebastiani P, Andersen SL, Sweigart B, Du M, Cosentino S, Thyagarajan B, Christensen K, Schupf N, Perls TT. Patterns of multi-domain cognitive aging in participants of the Long Life Family Study. GeroScience 2020; 42:1335-1350. [PMID: 32514870 PMCID: PMC7525612 DOI: 10.1007/s11357-020-00202-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 05/08/2020] [Indexed: 12/16/2022] Open
Abstract
Maintaining good cognitive function at older age is important, but our knowledge of patterns and predictors of cognitive aging is still limited. We used Bayesian model-based clustering to group 5064 participants of the Long Life Family Study (ages 49-110 years) into clusters characterized by distinct trajectories of cognitive change in the domains of episodic memory, attention, processing speed, and verbal fluency. For each domain, we identified 4 or 5 large clusters with representative patterns of change ranging from rapid decline to exceptionally slow change. We annotated the clusters by their correlation with genetic and molecular biomarkers, non-genetic risk factors, medical history, and other markers of aging to discover correlates of cognitive changes and neuroprotection. The annotation analysis discovered both predictors of multi-domain cognitive change such as gait speed and predictors of domain-specific cognitive change such as IL6 and NTproBNP that correlate only with change of processing speed or APOE genotypes that correlate only with change of processing speed and logical memory. These patterns also suggest that cognitive decline starts at young age and that maintaining good physical function correlates with slower cognitive decline. To better understand the agreement of cognitive changes across multiple domains, we summarized the results of the cluster analysis into a score of cognitive function change. This score showed that extreme patterns of change affecting multiple cognitive domains simultaneously are rare in this study and that specific signatures of biomarkers of inflammation and metabolic disease predict severity of cognitive changes. The substantial heterogeneity of change patterns within and between cognitive domains and the net of correlations between patterns of cognitive aging and other aging traits emphasizes the importance of measuring a wide range of cognitive functions and the need for studying cognitive aging in concert with other aging traits.
Collapse
Affiliation(s)
- Paola Sebastiani
- Department of Biostatistics, Boston University School of Public Health, 801 Massachusetts Avenue, Boston, MA 02118 USA
| | - Stacy L. Andersen
- Department of Medicine, Geriatrics Section, Boston University School of Medicine, Robinson 2400, 72 E Concord St, Boston, MA 02118 USA
| | - Benjamin Sweigart
- Department of Biostatistics, Boston University School of Public Health, 801 Massachusetts Avenue, Boston, MA 02118 USA
| | - Mengtian Du
- Department of Biostatistics, Boston University School of Public Health, 801 Massachusetts Avenue, Boston, MA 02118 USA
| | - Stephanie Cosentino
- Department of Neurology, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, and the Gertrude H. Sergievsky Center, Columbia University Medical Center, 630 West 168th Street, New York, NY 10032 USA
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, MMC 609 Mayo, 420 Delaware, Minneapolis, MN 55455 USA
| | - Kaare Christensen
- Department of Public Health, The Danish Aging Research Center and The Danish Twin Registry, Epidemiology, Biostatistics and Biodemography, University of Southern Denmark, 5000 Odense, Denmark
| | - Nicole Schupf
- Department of Neurology, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, and the Gertrude H. Sergievsky Center, Columbia University Medical Center, 630 West 168th Street, New York, NY 10032 USA
- Department of Epidemiology, Sergievsky Center, Columbia University Mailman School of Public Health, 630 West 168th Street, New York, NY 10032 USA
| | - Thomas T Perls
- Department of Medicine, Geriatrics Section, Boston University School of Medicine, Robinson 2400, 72 E Concord St, Boston, MA 02118 USA
| |
Collapse
|
44
|
LaSorda KR, Gmelin T, Kuipers AL, Boudreau RM, Santanasto AJ, Christensen K, Renner SW, Wojczynski MK, Andersen SL, Cosentino S, Glynn NW. Epidemiology of Perceived Physical Fatigability in Older Adults: The Long Life Family Study. J Gerontol A Biol Sci Med Sci 2020; 75:e81-e88. [PMID: 31828303 PMCID: PMC7494027 DOI: 10.1093/gerona/glz288] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Fatigability is a construct that measures whole-body tiredness anchored to activities of a fixed intensity and duration; little is known about its epidemiology and heritability. METHODS Two generations of family members enriched for exceptional longevity and their spouses were enrolled (2006-2009) in the Long Life Family Study (LLFS). At Visit 2 (2014-2017, N = 2,355) perceived physical fatigability was measured using the 10-item self-administered Pittsburgh Fatigability Scale (PFS), along with demographic, medical, behavioral, physical, and cognitive risk factors. RESULTS Residual genetic heritability of fatigability was 0.263 (p = 6.6 × 10-9) after adjustment for age, sex, and field center. PFS physical scores (mean ± SD) and higher physical fatigability prevalence (% PFS ≥ 15) were greater with each age strata: 60-69 (n = 1,009, 11.0 ± 7.6, 28%), 70-79 (n = 847, 12.5 ± 8.1, 37%), 80-89 (n = 253, 19.3 ± 9.9, 65.2%), and 90-108 (n = 266, 28.6 ± 9.8, 89.5%), p < .0001, adjusted for sex, field center, and family relatedness. Women had a higher prevalence of perceived physical fatigability compared to men, with the largest difference in the 80-89 age strata, 74.8% versus 53.5%, p < .0001. Those with greater body mass index, worse physical and cognitive function, and lower physical activity had significantly higher perceived physical fatigability. CONCLUSIONS Perceived physical fatigability is highly prevalent in older adults and strongly associated with age. The family design of LLFS allowed us to estimate the genetic heritability of perceived physical fatigability. Identifying risk factors associated with higher perceived physical fatigability can inform the development of targeted interventions for those most at risk, including older women, older adults with depression, and those who are less physically active.
Collapse
Affiliation(s)
- Kelsea R LaSorda
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Theresa Gmelin
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Allison L Kuipers
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Robert M Boudreau
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Adam J Santanasto
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kaare Christensen
- Department of Public Health, Unit of Epidemiology, Biostatistics and Biodemography, University of Southern Denmark, Odense, Denmark
| | - Sharon W Renner
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mary K Wojczynski
- Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | - Stacy L Andersen
- Geriatrics Section, Department of Medicine, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts
| | - Stephanie Cosentino
- Department of Neurology, Columbia University Medical Center, New York, New York
| | - Nancy W Glynn
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
45
|
Galvin A, Ukraintseva S, Arbeev K, Feitosa M, Christensen K. Physical robustness and resilience among long-lived female siblings: a comparison with sporadic long-livers. Aging (Albany NY) 2020; 12:15157-15168. [PMID: 32652515 PMCID: PMC7425496 DOI: 10.18632/aging.103618] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 06/22/2020] [Indexed: 11/25/2022]
Abstract
Long-lived individuals are central in studies of healthy longevity. However, few pro-longevity factors have been identified, presumably because of “phenocopies”, i.e. individuals that live long by chance. Familial longevity cases may include less phenocopies than sporadic cases and provide better insights into longevity mechanisms. Here we examined whether long-lived female siblings have a better ability to avoid diseases at ages 65+ (proxy for “robustness”) and/or survive to extreme ages (proxy for “resilience”) compared to sporadic long-livers. A total of 1,156 long-lived female siblings were selected from three nationwide Danish studies and age-matched with sporadic long-lived female controls. Outcomes included cumulative incidence of common health disorders from age 65 and overall survival. Long-lived female siblings had lower risks of some but not all health conditions, most significantly, depression (OR=0.74; 95%CI=0.62-0.88), and less significantly hypertensive (OR=0.84; 95%CI=0.71-0.99) and cerebrovascular (OR=0.73; 95%CI=0.55-0.96) diseases. They also had consistently better survival to extreme ages (HR=0.71; 95%CI= 0.63-0.81) compared to sporadic long-livers. After adjustment for the diseases, the association with mortality changed only marginally suggesting central role of better physiological resilience in familial longevity. Due to their consistently better resilience, familial longevity cases could be more informative than sporadic cases for studying mechanisms of healthy longevity.
Collapse
Affiliation(s)
- Angéline Galvin
- Epidemiology, Biostatistics, and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | | | - Konstantin Arbeev
- Center for Population Health and Aging, Duke University, Durham, NC 27708, USA
| | - Mary Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kaare Christensen
- Epidemiology, Biostatistics, and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark.,The Danish Aging Research Center, Department of Public Health, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
46
|
Gurinovich A, Andersen SL, Puca A, Atzmon G, Barzilai N, Sebastiani P. Varying Effects of APOE Alleles on Extreme Longevity in European Ethnicities. J Gerontol A Biol Sci Med Sci 2020; 74:S45-S51. [PMID: 31724059 DOI: 10.1093/gerona/glz179] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Indexed: 12/19/2022] Open
Abstract
APOE is a well-studied gene with multiple effects on aging and longevity. The gene has three alleles: e2, e3, and e4, whose frequencies vary by ethnicity. While the e2 is associated with healthy cognitive aging, the e4 allele is associated with Alzheimer's disease and early mortality and therefore its prevalence among people with extreme longevity (EL) is low. Using the PopCluster algorithm, we identified several ethnically different clusters in which the effect of the e2 and e4 alleles on EL changed substantially. For example, PopCluster discovered a large group of 1,309 subjects enriched of Southern Italian genetic ancestry with weaker protective effect of e2 (odds ratio [OR] = 1.27, p = .14) and weaker damaging effect of e4 (OR = 0.82, p = .31) on the phenotype of EL compared to other European ethnicities. Further analysis of this cluster suggests that the odds for EL in carriers of the e4 allele with Southern Italian genetic ancestry differ depending on whether they live in the United States (OR = 0.29, p = .009) or Italy (OR = 1.21, p = .38). PopCluster also found clusters enriched of subjects with Danish ancestry with varying effect of e2 on EL. The country of residence (Denmark or United States) appears to change the odds for EL in the e2 carriers.
Collapse
Affiliation(s)
- Anastasia Gurinovich
- Bioinformatics Program, Boston University, Massachusetts.,Department of Biostatistics, Boston University School of Public Health, Massachusetts
| | | | - Annibale Puca
- Department of Medicine and Surgery, University of Salerno, Fisciano, SA, Italy.,Cardiovascular Research Unit, IRCCS MultiMedica, Sesto San Giovanni, MI, Italy
| | - Gil Atzmon
- Faculty of Natural Science, University of Haifa, Israel.,Albert Einstein College of Medicine, Bronx, New York
| | - Nir Barzilai
- Albert Einstein College of Medicine, Bronx, New York
| | - Paola Sebastiani
- Department of Biostatistics, Boston University School of Public Health, Massachusetts
| |
Collapse
|
47
|
Berg N, Rodríguez‐Girondo M, Mandemakers K, Janssens AAPO, Beekman M, Slagboom PE. Longevity Relatives Count score identifies heritable longevity carriers and suggests case improvement in genetic studies. Aging Cell 2020; 19:e13139. [PMID: 32352215 PMCID: PMC7294789 DOI: 10.1111/acel.13139] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 01/24/2020] [Accepted: 02/23/2020] [Indexed: 12/23/2022] Open
Abstract
Loci associated with longevity are likely to harbor genes coding for key players of molecular pathways involved in a lifelong decreased mortality and decreased/compressed morbidity. However, identifying such loci is challenging. One of the most plausible reasons is the uncertainty in defining long‐lived cases with the heritable longevity trait among long‐living phenocopies. To avoid phenocopies, family selection scores have been constructed, but these have not yet been adopted as state of the art in longevity research. Here, we aim to identify individuals with the heritable longevity trait by using current insights and a novel family score based on these insights. We use a unique dataset connecting living study participants to their deceased ancestors covering 37,825 persons from 1,326 five‐generational families, living between 1788 and 2019. Our main finding suggests that longevity is transmitted for at least two subsequent generations only when at least 20% of all relatives are long‐lived. This proves the importance of family data to avoid phenocopies in genetic studies.
Collapse
Affiliation(s)
- Niels Berg
- Section of Molecular Epidemiology Department of Biomedical Data Sciences Leiden University Medical Center Leiden The Netherlands
- Radboud Group for Historical Demography and Family History Radboud University Nijmegen The Netherlands
| | - Mar Rodríguez‐Girondo
- Section of Medical Statistics Department of Biomedical Data Sciences Leiden University Medical Center Leiden The Netherlands
| | - Kees Mandemakers
- International Institute of Social History Amsterdam The Netherlands
| | | | - Marian Beekman
- Section of Molecular Epidemiology Department of Biomedical Data Sciences Leiden University Medical Center Leiden The Netherlands
| | - P. Eline Slagboom
- Section of Molecular Epidemiology Department of Biomedical Data Sciences Leiden University Medical Center Leiden The Netherlands
- Max Planck Institute for Biology of Ageing Cologne Germany
| |
Collapse
|
48
|
Santanasto AJ, Marron MM, Boudreau RM, Feitosa MF, Wojczynski MK, Arbeev KG, Thyagarajan B, Schupf N, Stallard E, Sebastiani P, Cosentino S, Christensen K, Newman AB. Prevalence, Incidence, and Risk Factors for Overall, Physical, and Cognitive Independence Among Those From Exceptionally Long-Lived Families: The Long Life Family Study. J Gerontol A Biol Sci Med Sci 2020; 75:899-905. [PMID: 31086986 PMCID: PMC7164521 DOI: 10.1093/gerona/glz124] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND The Long Life Family Study (LLFS) enrolled families exhibiting exceptional longevity. The goal of this article was to determine the prevalence and predictors of remaining independent after 7 years in the oldest generation. METHODS We examined 7-year change in physical (free of activities of daily living difficulty), cognitive (Mini-Mental State Examination score ≥ 24), and overall independence (physically/cognitively independent) in adults aged 90.3 ± 6.3 from LLFS's oldest generation. Potential predictors (n = 28) of remaining independent included demographics, diseases, biomarkers, anthropometrics, and physical and cognitive performance tasks and were determined using generalized estimating equations (α: p < .05). This was a discovery/exploratory analysis, so no multiple testing correction was employed and the results require independent replication. RESULTS At baseline (n = 1442), 67.3%, 83.8%, and 79.7% were overall, physically, and cognitively independent, respectively. After 7 years, 66% died, 7.5% were lost to follow-up, and the prevalence of overall independence decreased to 59.1% in survivors (-8.2%, 95% confidence interval: -14.1%, 2.2%). Of those with baseline independence, 156/226 (69.0%) remained independent. Predictors of remaining physically independent included younger age, better Short Physical Performance Battery score and lung function, smaller waist circumference, and lower soluble receptor for advanced glycation end-product levels (p < .05). Predictors of remaining cognitively independent included no cancer history, better Digit Symbol Substitution Test performance, and higher body weight (p < .05). CONCLUSIONS The prevalence of independence decreased by only 8.2% after 7 years, demonstrating the close correspondence between disability and mortality. Further, despite a mean baseline age of 90 years, a large proportion of survivors remained independent, suggesting this exceptional subgroup may harbor protective mechanisms.
Collapse
Affiliation(s)
- Adam J Santanasto
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pennsylvania
| | - Megan M Marron
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pennsylvania
| | - Robert M Boudreau
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pennsylvania
| | - Mary F Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine in St. Louis, Missouri
| | - Mary K Wojczynski
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine in St. Louis, Missouri
| | - Konstantin G Arbeev
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Nicole Schupf
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York
| | - Eric Stallard
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina
| | - Paola Sebastiani
- Department of Biostatistics, School of Public Health, Boston University, Massachusetts
| | - Stephanie Cosentino
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York
| | - Kaare Christensen
- Epidemiology Unit, Institute of Public Health, The Danish Aging Research Center, University of Southern Denmark, Sønderborg, Denmark
| | - Anne B Newman
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pennsylvania
| |
Collapse
|
49
|
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
|
50
|
Arbeev KG, Bagley O, Ukraintseva SV, Wu D, Duan H, Kulminski AM, Stallard E, Christensen K, Lee JH, Thyagarajan B, Zmuda JM, Yashin AI. Genetics of physiological dysregulation: findings from the long life family study using joint models. Aging (Albany NY) 2020; 12:5920-5947. [PMID: 32235003 PMCID: PMC7185144 DOI: 10.18632/aging.102987] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 03/24/2020] [Indexed: 12/16/2022]
Abstract
Recently, Mahalanobis distance (DM) was suggested as a statistical measure of physiological dysregulation in aging individuals. We constructed DM variants using sets of biomarkers collected at the two visits of the Long Life Family Study (LLFS) and performed joint analyses of longitudinal observations of DM and follow-up mortality in LLFS using joint models. We found that DM is significantly associated with mortality (hazard ratio per standard deviation: 1.31 [1.16, 1.48] to 2.22 [1.84, 2.67]) after controlling for age and other covariates. GWAS of random intercepts and slopes of DM estimated from joint models found a genome-wide significant SNP (rs12652543, p=7.2×10-9) in the TRIO gene associated with the slope of DM constructed from biomarkers declining in late life. Review of biological effects of genes corresponding to top SNPs from GWAS of DM slopes revealed that these genes are broadly involved in cancer prognosis and axon guidance/synapse function. Although axon growth is mainly observed during early development, the axon guidance genes can function in adults and contribute to maintenance of neural circuits and synaptic plasticity. Our results indicate that decline in axons' ability to maintain complex regulatory networks may potentially play an important role in the increase in physiological dysregulation during aging.
Collapse
Affiliation(s)
- Konstantin G Arbeev
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham NC, 27708, USA
| | - Olivia Bagley
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham NC, 27708, USA
| | - Svetlana V Ukraintseva
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham NC, 27708, USA
| | - Deqing Wu
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham NC, 27708, USA
| | - Hongzhe Duan
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham NC, 27708, USA
| | - Alexander M Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham NC, 27708, USA
| | - Eric Stallard
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham NC, 27708, USA
| | - Kaare Christensen
- Danish Aging Research Center, Department of Public Health, University of Southern Denmark 5000, Odense C, Denmark
| | - Joseph H Lee
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY 10032, USA.,G. H. Sergievsky Center, Columbia University, New York, NY 10032, USA.,Departments of Epidemiology and Neurology, Columbia University Medical Center, New York, NY 10032, USA
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Joseph M Zmuda
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Anatoliy I Yashin
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham NC, 27708, USA
| |
Collapse
|