1
|
Heikkinen A, Esser VFC, Lee SHT, Lundgren S, Hakkarainen A, Lundbom J, Kuula J, Groop PH, Heinonen S, Villicaña S, Bell JT, Maguolo A, Nilsson E, Ling C, Vaag A, Pajukanta P, Kaprio J, Pietiläinen KH, Li S, Ollikainen M. Twin pair analysis uncovers links between DNA methylation, mitochondrial DNA quantity and obesity. Nat Commun 2025; 16:4374. [PMID: 40355419 PMCID: PMC12069627 DOI: 10.1038/s41467-025-59576-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 04/24/2025] [Indexed: 05/14/2025] Open
Abstract
Alterations in mitochondrial metabolism in obesity may indicate disrupted communication between mitochondria and nucleus, and DNA methylation may influence this interplay. Here, we leverage data from the Finnish Twin Cohort study subcohort (n = 173; 86 full twin pairs, 1 singleton), including comprehensive measurements of obesity-related outcomes, mitochondrial DNA quantity and nuclear DNA methylation levels in adipose and muscle tissue, to identify one CpG at SH3BP4 significantly associated with mitochondrial DNA quantity in adipose tissue (FDR < 0.05). We also show that SH3BP4 methylation correlates with its gene expression. Additionally, we find that 14 out of the 35 obesity-related traits display significant associations with both SH3BP4 methylation and mitochondrial DNA quantity in adipose tissue. We use data from TwinsUK and the Scandinavian T2D-discordant monozygotic twin cohort, to validate the observed associations. Further analysis using ICE FALCON suggests that mitochondrial DNA quantity, insulin sensitivity and certain body fat measures are causal to SH3BP4 methylation. Examining mitochondrial DNA quantity and obesity-related traits suggests causation from mitochondrial DNA quantity to obesity, but unmeasured within-individual confounding cannot be ruled out. Our findings underscore the impact of mitochondrial DNA quantity on DNA methylation and expression of the SH3BP4 gene within adipose tissue, with potential implications for obesity.
Collapse
Affiliation(s)
- Aino Heikkinen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland.
- Minerva Foundation Institute for Medical Research, Helsinki, Finland.
| | - Vivienne F C Esser
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, VIC, Australia
| | - Seung Hyuk T Lee
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Sara Lundgren
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Antti Hakkarainen
- HUS Medical Imaging Center, Radiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Jesper Lundbom
- HUS Medical Imaging Center, Radiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University, Düsseldorf, Germany
| | - Juho Kuula
- HUS Medical Imaging Center, Radiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Public Health Promotion Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Sini Heinonen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Internal Medicine, Helsinki University Hospital, Helsinki, Finland
| | - Sergio Villicaña
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Jordana T Bell
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Alice Maguolo
- Epigenetics and Diabetes Unit, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| | - Emma Nilsson
- Epigenetics and Diabetes Unit, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| | - Charlotte Ling
- Epigenetics and Diabetes Unit, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| | - Allan Vaag
- Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
- Copenhagen University Hospital, Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Endocrinology, Skåne University Hospital, Malmö, Sweden
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, USA
- Institute for Precision Health, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- HealthyWeightHub, Endocrinology, Abdominal Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Shuai Li
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, VIC, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
| | - Miina Ollikainen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland.
- Minerva Foundation Institute for Medical Research, Helsinki, Finland.
| |
Collapse
|
2
|
Stephenson M, Barr P, Thomas N, Cooke M, Latvala A, Rose RJ, Kaprio J, Dick D, Salvatore JE. Patterns and predictors of alcohol misuse trajectories from adolescence through early midlife. Dev Psychopathol 2025; 37:734-750. [PMID: 38465371 PMCID: PMC11387953 DOI: 10.1017/s0954579424000543] [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] [Indexed: 03/12/2024]
Abstract
We took a multilevel developmental contextual approach and characterized trajectories of alcohol misuse from adolescence through early midlife, examined genetic and environmental contributions to individual differences in those trajectories, and identified adolescent and young adult factors associated with change in alcohol misuse. Data were from two longitudinal population-based studies. FinnTwin16 is a study of Finnish twins assessed at 16, 17, 18, 25, and 35 years (N = 5659; 52% female; 32% monozygotic). The National Longitudinal Study of Adolescent to Adult Health (Add Health) is a study of adolescents from the United States, who were assessed at five time points from 1994 to 2018 (N = 18026; 50% female; 64% White, 21% Black, 4% Native American, 7% Asian, 9% Other race/ethnicity). Alcohol misuse was measured as frequency of intoxication in FinnTwin16 and frequency of binge drinking in Add Health. In both samples, trajectories of alcohol misuse were best described by a quadratic growth curve: Alcohol misuse increased across adolescence, peaked in young adulthood, and declined into early midlife. Individual differences in these trajectories were primarily explained by environmental factors. Several adolescent and young adult correlates were related to the course of alcohol misuse, including other substance use, physical and mental health, and parenthood.
Collapse
Affiliation(s)
- Mallory Stephenson
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Peter Barr
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Nathaniel Thomas
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA
| | - Megan Cooke
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Antti Latvala
- Institute of Criminology and Legal Policy, University of Helsinki, Helsinki, Finland
| | - Richard J Rose
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Jaakko Kaprio
- Department of Public Health, Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Danielle Dick
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Jessica E Salvatore
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| |
Collapse
|
3
|
Muniandy M, Joenväärä S, van der Kolk BW, Tohmola T, Haltia H, Saari S, Hakkarainen A, Lundbom J, Kuula J, Groop PH, Kaprio J, Heinonen S, Renkonen R, Pietiläinen KH. Plasma N-Glycoproteomics in monozygotic twin pairs discordant for body mass index reveals an obesity signature related to inflammation and iron metabolism. Biol Direct 2025; 20:31. [PMID: 40108677 PMCID: PMC11921541 DOI: 10.1186/s13062-025-00609-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 01/20/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND N-glycosylation is a complex, post-translational modification which influences protein function and is sensitive to physiological changes. Obesity is associated with alterations in protein function; however, little is known about the glycoproteome in obesity beyond observations of association with types and structures of selected glycopeptides. Most often, due to technical challenges, glycan composition and structure information are missing. Here, we combined label-free data-independent proteomics and targeted quantitative glycoproteomics to study N-glycosylation of plasma proteins in obesity. Using a monozygotic twin study design, we controlled for genetic variation and focused only on the acquired effects of obesity. METHODS Using plasma samples of 48 monozygotic twin pairs discordant for BMI (intrapair difference > 2.5 kg/m2), we identified using mass spectrometry, differential protein and glycopeptide levels between heavier and leaner co-twins. We used a within-twin paired analysis model and considered p < 0.05 as significant. RESULTS We identified 48 protein and 33 N-glycosylation expression differences (p < 0.05) between co-twins. These differences occurred either both in the protein expression and glycoprotein (sometimes in opposing directions) or independently from each other. Haptoglobin protein was upregulated (Fold Change = 1.10, p = 0.001) in heavier co-twins along with seven upregulated glycan compositions at N-glycosylation site Asn241. The complement protein C3 was upregulated (Fold Change = 1.08, p = 0.014) along with one upregulated glycopeptide at Asn85. Additionally, many glycopeptides were upregulated despite non-significant differences in protein-backbone plasma levels. CONCLUSION Differential protein expression related to cholesterol biosynthesis and acute phase signalling as well as N-glycosylation of proteins related to iron metabolism and inflammation can be linked to acquired obesity.
Collapse
Affiliation(s)
- Maheswary Muniandy
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Sakari Joenväärä
- Transplantation Laboratory, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Birgitta W van der Kolk
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Tiialotta Tohmola
- Transplantation Laboratory, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Hanna Haltia
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sina Saari
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Antti Hakkarainen
- Department of Radiology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Jesper Lundbom
- Department of Radiology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University, Düsseldorf, Germany
| | - Juho Kuula
- Department of Radiology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
- Faculty of Medicine Doctoral Program in Clinical Research, University of Helsinki, Helsinki, Finland
| | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
| | - Sini Heinonen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Risto Renkonen
- Transplantation Laboratory, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Healthy Weight Hub, Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
4
|
Cooke ME, Lumpe E, Stephenson M, Urjansson M, Aliev F, Palviainen T, Brislin SJ, Piirtola M, Rabinowitz J, Latvala A, Barr PB, Vuoksimaa E, Maes HHM, Viken R, Rose RJ, Kaprio J, Dick DM, Aaltonen S, Salvatore JE. Alcohol use in Early Midlife: Findings from the Age 37 Follow-Up Assessment of the FinnTwin12 Cohort. Behav Genet 2025; 55:124-140. [PMID: 39920525 PMCID: PMC11882652 DOI: 10.1007/s10519-024-10212-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 12/31/2024] [Indexed: 02/09/2025]
Abstract
This paper provides an overview of the most recent assessment, collected in early midlife, of the FinnTwin12 cohort, a population-based study of Finnish twins born in 1983-1987. The twins were invited to complete an online survey assessing a range of variables, including physical and mental health, alcohol use and problems, other substance use, and early midlife environments (e.g., parenthood). In total, 2,085 individuals (~ 40% of the original sample) completed the survey (551 complete twin pairs, 58.7% female, 37.3% monozygotic, Mage = 37.2 years, SD = 1.47 years, age range = 34-39 years). Individuals who participated were more likely to be female, monozygotic, and have higher parental education and less hyperactivity/impulsivity and aggression at age 12 when compared to individuals who were invited but did not participate. Parental alcohol misuse and the twins' alcohol use and misuse at age 14 were not related to study retention. Alcohol misuse in early midlife was positively associated with nicotine dependence, lifetime use of cannabis and other drugs, trauma exposure, and depressive symptoms, and negatively associated with physical health and having biological children. These new data expand upon the wealth of measures collected as part of previous assessments, expanding the scope of work on the etiology and correlates of alcohol misuse within a longitudinal, genetically-informed framework. In addition to these new survey measures, we are planning an in-person assessment to collect physiological measurements and conduct additional in-depth phenotyping on a subset of twins who have been more intensively studied over the years.
Collapse
Affiliation(s)
- Megan E Cooke
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, USA
| | - Erin Lumpe
- Department of Psychology, Rutgers University, New Brunswick, USA
| | - Mallory Stephenson
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, USA
| | - Mia Urjansson
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Fazil Aliev
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, USA
| | - Teemu Palviainen
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Sarah J Brislin
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, USA
| | - Maarit Piirtola
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
- UKK Institute for Health Promotion Research, Tampere, Finland
| | - Jill Rabinowitz
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, USA
| | - Antti Latvala
- Institute of Criminology and Legal Policy, University of Helsinki, Helsinki, Finland
| | - Peter B Barr
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, USA
| | - Eero Vuoksimaa
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Hermine H M Maes
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, USA
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, USA
- Department of Psychiatry, Virginia Commonwealth University, Richmond, USA
- Massey Cancer Center, Virginia Commonwealth University, Richmond, USA
| | - Richard Viken
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, USA
| | - Richard J Rose
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, USA
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland.
| | - Danielle M Dick
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, USA.
| | - Sari Aaltonen
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland.
| | - Jessica E Salvatore
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, USA.
| |
Collapse
|
5
|
Lumpe E, Pascale A, Stephenson M, Barr P, Cooke ME, Latvala A, Maes HHM, Aaltonen S, Piirtola M, Viken R, Rose RJ, Sipilä PN, Keski‐Rahkonen A, Vuoksimaa E, Kaprio J, Dick DM, Salvatore JE. Etiology and correlates of alcohol misuse in early midlife. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2025; 49:301-314. [PMID: 39704247 PMCID: PMC11828966 DOI: 10.1111/acer.15513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 12/02/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND Early midlife individuals (ages 30-40) experience demographic shifts that may influence the remainder of adult life. Although new or persistent alcohol misuse is common during this period, early midlife is understudied in alcohol use literature. We examined the heritability of alcohol misuse; the associations between alcohol misuse and sociodemographic factors, physical health, and well-being; and whether these associations were robust in cotwin comparisons. METHODS Participants were 1446 Finnish twin pairs and 748 nonpaired Finnish twins with mean age 34 years. The alcohol misuse index was a composite measure of frequency of use, intoxication, heavy episodic drinking, and alcohol problems assessed with the Malmö-modified Michigan Alcoholism Screening Test and the Rutgers Alcohol Problem Index. Early midlife correlates included relationship status and length, family formation, unemployment status, education level, self-rated health, pain, sleeping difficulties, life satisfaction, psychological health, and other substance use. We employed a sex-limitation model to estimate early midlife heritability. Linear and fixed effects regression models were used for individual and cotwin comparison analyses, respectively. RESULTS Additive genetic (A) and unique environmental (E) components of alcohol misuse variance differed across sex (Females: A = 62%, E = 38%; Males: A = 49%, E = 51%). In individual-based analyses, higher scores on the alcohol misuse index were associated with lower relationship stability, financial situation, education level, self-rated health, physical fitness, life satisfaction and psychological health, and higher self-reported pain, sleep difficulties, unemployment rates and other substance use (R2 = 0.008-0.12). Associations remained significant in cotwin comparison analyses (R2 = 0.004-0.10) except for financial situation and education level. CONCLUSIONS There is evidence of sex differences in the etiological factors that influence early midlife drinking. After controlling for confounding familial factors, associations between alcohol misuse and poorer early midlife functioning largely remained, suggesting that alcohol misuse may play a role in poorer functioning across several outcomes.
Collapse
Affiliation(s)
- Erin Lumpe
- Department of PsychologyRutgers UniversityNew BrunswickNew JerseyUSA
| | - Angela Pascale
- Department of PsychologyVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Mallory Stephenson
- Department of PsychologyVirginia Commonwealth UniversityRichmondVirginiaUSA
- Virginia Institute for Psychiatric and Behavioral GeneticsRichmondVirginiaUSA
| | - Peter Barr
- Department of Psychiatry and Behavioral SciencesSUNY Downstate Health Sciences UniversityBrooklynNew YorkUSA
| | - Megan E. Cooke
- Department of PsychiatryRutgers Robert Wood Johnson School of MedicinePiscatawayNew JerseyUSA
| | - Antti Latvala
- Institute of Criminology and Legal PolicyUniversity of HelsinkiHelsinkiFinland
| | - Hermine H. M. Maes
- Virginia Institute for Psychiatric and Behavioral GeneticsRichmondVirginiaUSA
| | - Sari Aaltonen
- Institute for Molecular Medicine Finland (FIMM)University of HelsinkiHelsinkiFinland
| | - Maarit Piirtola
- Institute for Molecular Medicine Finland (FIMM)University of HelsinkiHelsinkiFinland
- UKK Institute for Health Promotion ResearchTampereFinland
| | - Richard Viken
- Department of Psychological and Brain SciencesIndiana UniversityBloomingtonIndianaUSA
| | - Richard J. Rose
- Department of Psychological and Brain SciencesIndiana UniversityBloomingtonIndianaUSA
| | - Pyry N. Sipilä
- Department of Public HealthUniversity of HelsinkiHelsinkiFinland
| | | | - Eero Vuoksimaa
- Institute for Molecular Medicine Finland (FIMM)University of HelsinkiHelsinkiFinland
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland (FIMM)University of HelsinkiHelsinkiFinland
| | - Danielle M. Dick
- Rutgers Addiction Research CenterRutgers Brain Health InstitutePiscatawayNew JerseyUSA
| | - Jessica E. Salvatore
- Department of PsychiatryRutgers Robert Wood Johnson School of MedicinePiscatawayNew JerseyUSA
| |
Collapse
|
6
|
Obeso A, Drouard G, Jelenkovic A, Aaltonen S, Palviainen T, Salvatore JE, Dick DM, Kaprio J, Silventoinen K. Genetic contributions to body mass index over adolescence and its associations with adult weight gain: a 25-year follow-up study of Finnish twins. Int J Obes (Lond) 2025; 49:357-363. [PMID: 39567637 PMCID: PMC11805703 DOI: 10.1038/s41366-024-01684-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024]
Abstract
INTRODUCTION High body mass index (BMI) in adolescence is a strong predictor of adult obesity. However, the nature of this association is unclear. We investigated how adolescent BMI is associated with adult weight change using longitudinal data from ages 11.5 to 37 years and examined the genetic factors behind these associations. DATA AND METHODS The study cohort consisted of 1400 Finnish twin individuals (40% males) with 494 complete twin pairs who reported their body mass index (BMI) at five ages: 11.5, 14, 17.5, 24, and 37 years. BMI trajectories (defined as BMI changes (i.e., slope) and BMI at baseline age (i.e., intercept)) were calculated in adulthood (from 17.5 to 37 years of age) using linear mixed-effects models. Polygenic Risk Scores of BMI (PRSBMI) and genetic twin models were utilised to analyse the role of genetic factors underlying BMI trajectories and their associations with BMI at 11.5 and 14 years of age. RESULTS Mean BMI increased in adulthood (4.06 kg/m2 in men and 3.39 kg/m2 in women). The BMI changes correlated with BMI at the baseline age of 17.5 years (i.e. intercept) (r = 0.24 in men and r = 0.35 in women) as well as with BMI in adolescence (11.5 and 14 years of age). Genetic factors contributed to the BMI changes during adulthood (correlation with PRSBMI r = 0.25 in men and r = 0.27 in women; heritability estimates 0.63 and 0.64 respectively) as well as to their correlations with BMI at the baseline age (rA = 0.5 in men and 0.54 in women) and BMI during adolescence (at 11.5 and 14 years of age) (rA = 0.63-0.64). CONCLUSION We found that genetic factors play a role in BMI change in adulthood, and part of this genetic component overlaps with the genetics of BMI in adolescence. Genetic predisposition to high BMI in adolescence is also related to adult weight gain.
Collapse
Affiliation(s)
- Alvaro Obeso
- Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Science and Technology, University of the Basque Country, Bilbao, Spain.
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland.
| | - Gabin Drouard
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Aline Jelenkovic
- Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Science and Technology, University of the Basque Country, Bilbao, Spain
| | - Sari Aaltonen
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Teemu Palviainen
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Jessica E Salvatore
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Danielle M Dick
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Karri Silventoinen
- Helsinki Institute for Demography and Population Health, University of Helsinki, Helsinki, Finland
| |
Collapse
|
7
|
Deal M, Kar A, Lee SHT, Alvarez M, Rajkumar S, Arasu UT, Kaminska D, Männistö V, Heinonen S, van der Kolk BW, Säiläkivi U, Saarinen T, Juuti A, Pihlajamäki J, Kaikkonen MU, Laakso M, Pietiläinen KH, Pajukanta P. An abdominal obesity missense variant in the adipocyte thermogenesis gene TBX15 is implicated in adaptation to cold in Finns. Am J Hum Genet 2024; 111:2542-2560. [PMID: 39515300 PMCID: PMC11568758 DOI: 10.1016/j.ajhg.2024.10.001] [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: 05/22/2024] [Revised: 09/26/2024] [Accepted: 10/01/2024] [Indexed: 11/16/2024] Open
Abstract
Mechanisms of abdominal obesity GWAS variants have remained largely unknown. To elucidate these mechanisms, we leveraged subcutaneous adipose tissue (SAT) single nucleus RNA-sequencing and genomics data. After discovering that heritability of abdominal obesity is enriched in adipocytes, we focused on a SAT unique adipocyte marker gene, the transcription factor TBX15, and its abdominal obesity-associated deleterious missense variant, rs10494217. The allele frequency of rs10494217 revealed a north-to-south decreasing gradient, with consistent significant FST values observed for 25 different populations when compared to Finns, a population with a history of genetic isolation. Given the role of Tbx15 in mouse thermogenesis, the frequency may have increased as an adaptation to cold in Finns. Our selection analysis provided significant evidence of selection for the abdominal obesity risk allele T of rs10494217 in Finns, with a north-to-south decreasing trend in other populations, and demonstrated that latitude significantly predicts the allele frequency. We also discovered that the risk allele status significantly affects SAT adipocyte expression of multiple adipocyte marker genes in trans in two cohorts. Two of these trans genes have been connected to thermogenesis, supporting the thermogenic effect of the TBX15 missense variant as a possible cause of its selection. Adipose expression of one trans gene, a lncRNA, AC002066.1, was strongly associated with adipocyte size, implicating it in metabolically unhealthy adipocyte hypertrophy. In summary, the abdominal obesity variant rs10494217 was selected in Finns, and individuals with the risk allele have trans effects on adipocyte expression of genes relating to thermogenesis and adipocyte hypertrophy.
Collapse
Affiliation(s)
- Milena Deal
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Asha Kar
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA; Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, USA
| | - Seung Hyuk T Lee
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Marcus Alvarez
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sandhya Rajkumar
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Uma Thanigai Arasu
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Dorota Kaminska
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Department of Medicine, Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Ville Männistö
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Sini Heinonen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Birgitta W van der Kolk
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Ulla Säiläkivi
- Department of Abdominal Surgery, Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Tuure Saarinen
- Department of Abdominal Surgery, Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Anne Juuti
- Department of Abdominal Surgery, Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Jussi Pihlajamäki
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, Kuopio, Finland
| | - Minna U Kaikkonen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; HealthyWeightHub, Endocrinology, Abdominal Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Päivi Pajukanta
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA; Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, USA; Institute for Precision Health, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| |
Collapse
|
8
|
Zellers S, van Dongen J, Maes HHM, Ollikainen M, Fang F, Vrieze S, Kaprio J, Boomsma DI. A Bivariate Twin Study of Lifetime cannabis Initiation and Lifetime Regular Tobacco Smoking Across Three Different Countries. Behav Genet 2024; 54:375-385. [PMID: 39078541 PMCID: PMC11371858 DOI: 10.1007/s10519-024-10190-1] [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: 05/14/2024] [Accepted: 07/23/2024] [Indexed: 07/31/2024]
Abstract
Regular cigarette smoking and cannabis consumption are strongly positively related to each other, yet few studies explore their underlying variation and covariation. We evaluated the genetic and environmental decomposition of variance and covariance of these two traits in twin data from three countries with different social norms and legislation. Data from the Netherlands Twin Register, FinnTwin12/16, and the Minnesota Center for Twin Family Research (total N = 21,617) were analyzed in bivariate threshold models of lifetime regular smoking initiation (RSI) and lifetime cannabis initiation (CI). We ran unstratified models and models stratified by sex and country. Prevalence of RSI was lowest in the Netherlands and prevalence of CI was highest in Minnesota. In the unstratified model, genetic (A) and common environmental factors (C) contributed substantially to the liabilities of RSI (A = 0.47, C = 0.34) and CI (A = 0.28, C = 0.51). The two liabilities were significantly phenotypically (rP = 0.56), genetically (rA = 0.74), and environmentally correlated in the unstratified model (rC = 0.47and rE = 0.48, representing correlations between common and unique environmental factors). The magnitude of phenotypic correlation between liabilities varied by country but not sex (Minnesota rP ~ 0.70, Netherlands rP ~ 0.59, Finland rP ~ 0.45). Comparisons of decomposed correlations could not be reliably tested in the stratified models. The prevalence and association of RSI and CI vary by sex and country. These two behaviors are correlated because there is genetic and environmental overlap between their underlying latent liabilities. There is heterogeneity in the genetic architecture of these traits across country.
Collapse
Affiliation(s)
- Stephanie Zellers
- Institute for Molecular Medicine Finland, University of Helsinki, P.O. Box 20, Helsinki, 00014, Finland.
| | - Jenny van Dongen
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
| | - Hermine H M Maes
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Human and Molecular Genetics, Psychiatry and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Miina Ollikainen
- Institute for Molecular Medicine Finland, University of Helsinki, P.O. Box 20, Helsinki, 00014, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Fang Fang
- GenOmics and Translational Research Center, Research Triangle Institute International, Research Triangle Park, NC, USA
| | - Scott Vrieze
- Department of Psychology, University of Minnesota, Minneapolis, MN, USA
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland, University of Helsinki, P.O. Box 20, Helsinki, 00014, Finland
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
| |
Collapse
|
9
|
Föhr T, Hendrix A, Kankaanpää A, Laakkonen EK, Kujala U, Pietiläinen KH, Lehtimäki T, Kähönen M, Raitakari O, Wang X, Kaprio J, Ollikainen M, Sillanpää E. Metabolic syndrome and epigenetic aging: a twin study. Int J Obes (Lond) 2024; 48:778-787. [PMID: 38273034 PMCID: PMC11129944 DOI: 10.1038/s41366-024-01466-x] [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] [Received: 06/30/2023] [Revised: 12/13/2023] [Accepted: 01/10/2024] [Indexed: 01/27/2024]
Abstract
BACKGROUND Metabolic syndrome (MetS) is associated with premature aging, but whether this association is driven by genetic or lifestyle factors remains unclear. METHODS Two independent discovery cohorts, consisting of twins and unrelated individuals, were examined (N = 268, aged 23-69 years). The findings were replicated in two cohorts from the same base population. One consisted of unrelated individuals (N = 1 564), and the other of twins (N = 293). Participants' epigenetic age, estimated using blood DNA methylation data, was determined using the epigenetic clocks GrimAge and DunedinPACE. The individual-level linear regression models for investigating the associations of MetS and its components with epigenetic aging were followed by within-twin-pair analyses using fixed-effects regression models to account for genetic factors. RESULTS In individual-level analyses, GrimAge age acceleration was higher among participants with MetS (N = 56) compared to participants without MetS (N = 212) (mean 2.078 [95% CI = 0.996,3.160] years vs. -0.549 [-1.053,-0.045] years, between-group p = 3.5E-5). Likewise, the DunedinPACE estimate was higher among the participants with MetS compared to the participants without MetS (1.032 [1.002,1.063] years/calendar year vs. 0.911 [0.896,0.927] years/calendar year, p = 4.8E-11). An adverse profile in terms of specific MetS components was associated with accelerated aging. However, adjustments for lifestyle attenuated these associations; nevertheless, for DunedinPACE, they remained statistically significant. The within-twin-pair analyses suggested that genetics explains these associations fully for GrimAge and partly for DunedinPACE. The replication analyses provided additional evidence that the association between MetS components and accelerated aging is independent of the lifestyle factors considered in this study, however, suggesting that genetics is a significant confounder in this association. CONCLUSIONS The results of this study suggests that MetS is associated with accelerated epigenetic aging, independent of physical activity, smoking or alcohol consumption, and that the association may be explained by genetics.
Collapse
Affiliation(s)
- Tiina Föhr
- Faculty of Sport and Health Sciences, Gerontology Research Center, University of Jyväskylä, Jyväskylä, Finland.
| | - Arne Hendrix
- Physical Activity, Sport & Health Research Group, Department of Movement Sciences, KU Leuven - University of Leuven, Leuven, Belgium
| | - Anna Kankaanpää
- Faculty of Sport and Health Sciences, Gerontology Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - Eija K Laakkonen
- Faculty of Sport and Health Sciences, Gerontology Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - Urho Kujala
- Faculty of Sport and Health Sciences, Gerontology Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Healthy Weight Hub, Endocrinology, Abdominal Center, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, and Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Olli Raitakari
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| | - Xiaoling Wang
- Georgia Prevention Institute (GPI), Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Miina Ollikainen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Elina Sillanpää
- Faculty of Sport and Health Sciences, Gerontology Research Center, University of Jyväskylä, Jyväskylä, Finland
- The Wellbeing Services County of Central Finland, Jyväskylä, Finland
| |
Collapse
|
10
|
Romero Villela PN, Evans LM, Palviainen T, Border R, Kaprio J, Palmer RHC, Keller MC, Ehringer MA. Loci on chromosome 20 interact with rs16969968 to influence cigarettes per day in European ancestry individuals. Drug Alcohol Depend 2024; 257:111126. [PMID: 38387257 PMCID: PMC11062023 DOI: 10.1016/j.drugalcdep.2024.111126] [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] [Received: 10/20/2023] [Revised: 01/12/2024] [Accepted: 02/07/2024] [Indexed: 02/24/2024]
Abstract
BACKGROUND The understanding of the molecular genetic contributions to smoking is largely limited to the additive effects of individual single nucleotide polymorphisms (SNPs), but the underlying genetic risk is likely to also include dominance, epistatic, and gene-environment interactions. METHODS To begin to address this complexity, we attempted to identify genetic interactions between rs16969968, the most replicated SNP associated with smoking quantity, and all SNPs and genes across the genome. RESULTS Using the UK Biobank European subsample, we found one SNP, rs1892967, and two genes, PCNA and TMEM230, that showed a significant genome-wide interaction with rs16969968 for log10 CPD and raw CPD, respectively, in a sample of 116 442 individuals who self-reported currently or previously smoking. We extended these analyses to individuals of South Asian descent and meta-analyzed the combined sample of 117 212 individuals of European and South Asian ancestry. We replicated the gene findings in a meta-analysis of five Finnish samples (N=40 140): FinHealth, FINRISK, Finnish Twin Cohort, GeneRISK, and Health-2000-2011. CONCLUSIONS To our knowledge, this represents the first reliable epistatic association between single nucleotide polymorphisms for smoking behaviors and provides a novel direction for possible future functional studies related to this interaction. Furthermore, this work demonstrates the feasibility of these analyses by pooling multiple datasets across various ancestries, which may be applied to other top SNPs for smoking and/or other phenotypes.
Collapse
Affiliation(s)
- Pamela N Romero Villela
- Institute for Behavioral Genetics, University of Colorado, Boulder, USA; Department of Psychology and Neuroscience, University of Colorado, Boulder, USA
| | - Luke M Evans
- Institute for Behavioral Genetics, University of Colorado, Boulder, USA; Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, USA
| | - Teemu Palviainen
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, USA
| | - Richard Border
- Departments of Neurology and Computer Science, University of California, Los Angeles, USA
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, USA
| | - Rohan H C Palmer
- Behavioral Genetics of Addiction Laboratory, Department of Psychology, Emory University, Atlanta, GA, USA
| | - Matthew C Keller
- Institute for Behavioral Genetics, University of Colorado, Boulder, USA; Department of Psychology and Neuroscience, University of Colorado, Boulder, USA
| | - Marissa A Ehringer
- Institute for Behavioral Genetics, University of Colorado, Boulder, USA; Departments of Neurology and Computer Science, University of California, Los Angeles, USA; Department of Integrative Physiology, University of Colorado, Boulder, USA.
| |
Collapse
|
11
|
Elovainio M, Komulainen K, Hakulinen C, Pahkala K, Rovio S, Hutri N, Raitakari OT, Pulkki-Råback L. Intergenerational continuity of loneliness and potential mechanisms: Young Finns Multigenerational Study. Sci Rep 2024; 14:5465. [PMID: 38443584 PMCID: PMC10915156 DOI: 10.1038/s41598-024-56147-6] [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: 12/11/2023] [Accepted: 03/01/2024] [Indexed: 03/07/2024] Open
Abstract
Evidence on the intergenerational continuity of loneliness and on potential mechanisms that connect loneliness across successive generations is limited. We examined the association between loneliness of (G0) parents (859 mothers and 570 fathers, mean age 74 years) and their children (G1) (433 sons and 558 daughters, mean age 47 years) producing 991 parent-offspring pairs and tested whether these associations were mediated through subjective socioeconomic position, temperament characteristics, cognitive performance, and depressive symptoms. Mean loneliness across parents had an independent effect on their adult children's experienced loneliness (OR = 1.72, 95% CI 1.23-2.42). We also found a robust effect of mothers' (OR = 1.64, 95% CI 1.17-2.29), but not of fathers' loneliness (OR = 1.47, 95% CI 0.96-2.25) on offspring's experienced loneliness in adulthood. The associations were partly mediated by offspring depressive (41-54%) and anxiety (29-31%) symptoms. The current findings emphasize the high interdependence of loneliness within families mediated partly by offspring's mental health problems.
Collapse
Affiliation(s)
- Marko Elovainio
- Research Program Unit, Faculty of Medicine (Department of Psychology), University of Helsinki, Haartmaninkatu 3, P.O.Box 63, 00014, Helsinki, Finland.
- Finnish Institute for Health and Welfare, Mannerheimintie 166, 00300, Helsinki, Finland.
| | - Kaisla Komulainen
- Research Program Unit, Faculty of Medicine (Department of Psychology), University of Helsinki, Haartmaninkatu 3, P.O.Box 63, 00014, Helsinki, Finland
| | - Christian Hakulinen
- Research Program Unit, Faculty of Medicine (Department of Psychology), University of Helsinki, Haartmaninkatu 3, P.O.Box 63, 00014, Helsinki, Finland
| | - Katja Pahkala
- Department of Public Health, University of Turku and Turku University Hospital, Kiinamyllynkatu 10, 20520, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Kiinamyllynkatu 10, 20520, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland
| | - Suvi Rovio
- Department of Public Health, University of Turku and Turku University Hospital, Kiinamyllynkatu 10, 20520, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Kiinamyllynkatu 10, 20520, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland
| | - Nina Hutri
- Department of Pediatrics, Tampere University Hospital and Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Olli T Raitakari
- Department of Public Health, University of Turku and Turku University Hospital, Kiinamyllynkatu 10, 20520, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Kiinamyllynkatu 10, 20520, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland
| | - Laura Pulkki-Råback
- Research Program Unit, Faculty of Medicine (Department of Psychology), University of Helsinki, Haartmaninkatu 3, P.O.Box 63, 00014, Helsinki, Finland
| |
Collapse
|
12
|
Kar A, Alvarez M, Garske KM, Huang H, Lee SHT, Deal M, Das SS, Koka A, Jamal Z, Mohlke KL, Laakso M, Heinonen S, Pietiläinen KH, Pajukanta P. Age-dependent genes in adipose stem and precursor cells affect regulation of fat cell differentiation and link aging to obesity via cellular and genetic interactions. Genome Med 2024; 16:19. [PMID: 38297378 PMCID: PMC10829214 DOI: 10.1186/s13073-024-01291-x] [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: 09/11/2023] [Accepted: 01/19/2024] [Indexed: 02/02/2024] Open
Abstract
BACKGROUND Age and obesity are dominant risk factors for several common cardiometabolic disorders, and both are known to impair adipose tissue function. However, the underlying cellular and genetic factors linking aging and obesity on adipose tissue function have remained elusive. Adipose stem and precursor cells (ASPCs) are an understudied, yet crucial adipose cell type due to their deterministic adipocyte differentiation potential, which impacts the capacity to store fat in a metabolically healthy manner. METHODS We integrated subcutaneous adipose tissue (SAT) bulk (n=435) and large single-nucleus RNA sequencing (n=105) data with the UK Biobank (UKB) (n=391,701) data to study age-obesity interactions originating from ASPCs by performing cell-type decomposition, differential expression testing, cell-cell communication analyses, and construction of polygenic risk scores for body mass index (BMI). RESULTS We found that the SAT ASPC proportions significantly decrease with age in an obesity-dependent way consistently in two independent cohorts, both showing that the age dependency of ASPC proportions is abolished by obesity. We further identified 76 genes (72 SAT ASPC marker genes and 4 transcription factors regulating ASPC marker genes) that are differentially expressed by age in SAT and functionally enriched for developmental processes and adipocyte differentiation (i.e., adipogenesis). The 76 age-perturbed ASPC genes include multiple negative regulators of adipogenesis, such as RORA, SMAD3, TWIST2, and ZNF521, form tight clusters of longitudinally co-expressed genes during human adipogenesis, and show age-based differences in cellular interactions between ASPCs and adipose cell types. Finally, our genetic data demonstrate that cis-regional variants of these genes interact with age as predictors of BMI in an obesity-dependent way in the large UKB, while no such gene-age interaction on BMI is observed with non-age-dependent ASPC marker genes, thus independently confirming our cellular ASPC results at the biobank level. CONCLUSIONS Overall, we discover that obesity prematurely induces a decrease in ASPC proportions and identify 76 developmentally important ASPC genes that implicate altered negative regulation of fat cell differentiation as a mechanism for aging and directly link aging to obesity via significant cellular and genetic interactions.
Collapse
Affiliation(s)
- Asha Kar
- Department of Human Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles (UCLA), Gonda Center, Room 6357B, 695 Charles E. Young Drive South, Los Angeles, CA, 90095-7088, USA
| | - Marcus Alvarez
- Department of Human Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles (UCLA), Gonda Center, Room 6357B, 695 Charles E. Young Drive South, Los Angeles, CA, 90095-7088, USA
| | - Kristina M Garske
- Department of Human Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles (UCLA), Gonda Center, Room 6357B, 695 Charles E. Young Drive South, Los Angeles, CA, 90095-7088, USA
| | - Huiling Huang
- Department of Human Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles (UCLA), Gonda Center, Room 6357B, 695 Charles E. Young Drive South, Los Angeles, CA, 90095-7088, USA
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, USA
| | - Seung Hyuk T Lee
- Department of Human Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles (UCLA), Gonda Center, Room 6357B, 695 Charles E. Young Drive South, Los Angeles, CA, 90095-7088, USA
| | - Milena Deal
- Department of Human Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles (UCLA), Gonda Center, Room 6357B, 695 Charles E. Young Drive South, Los Angeles, CA, 90095-7088, USA
| | - Sankha Subhra Das
- Department of Human Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles (UCLA), Gonda Center, Room 6357B, 695 Charles E. Young Drive South, Los Angeles, CA, 90095-7088, USA
| | - Amogha Koka
- Department of Human Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles (UCLA), Gonda Center, Room 6357B, 695 Charles E. Young Drive South, Los Angeles, CA, 90095-7088, USA
| | - Zoeb Jamal
- Department of Human Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles (UCLA), Gonda Center, Room 6357B, 695 Charles E. Young Drive South, Los Angeles, CA, 90095-7088, USA
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Sini Heinonen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- HealthyWeightHub, Endocrinology, Abdominal Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLA, University of California, Los Angeles (UCLA), Gonda Center, Room 6357B, 695 Charles E. Young Drive South, Los Angeles, CA, 90095-7088, USA.
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, USA.
- Institute for Precision Health, David Geffen School of Medicine at UCLA, Los Angeles, USA.
| |
Collapse
|
13
|
Mbarek H, Gordon SD, Duffy DL, Hubers N, Mortlock S, Beck JJ, Hottenga JJ, Pool R, Dolan CV, Actkins KV, Gerring ZF, Van Dongen J, Ehli EA, Iacono WG, Mcgue M, Chasman DI, Gallagher CS, Schilit SLP, Morton CC, Paré G, Willemsen G, Whiteman DC, Olsen CM, Derom C, Vlietinck R, Gudbjartsson D, Cannon-Albright L, Krapohl E, Plomin R, Magnusson PKE, Pedersen NL, Hysi P, Mangino M, Spector TD, Palviainen T, Milaneschi Y, Penninnx BW, Campos AI, Ong KK, Perry JRB, Lambalk CB, Kaprio J, Ólafsson Í, Duroure K, Revenu C, Rentería ME, Yengo L, Davis L, Derks EM, Medland SE, Stefansson H, Stefansson K, Del Bene F, Reversade B, Montgomery GW, Boomsma DI, Martin NG. Genome-wide association study meta-analysis of dizygotic twinning illuminates genetic regulation of female fecundity. Hum Reprod 2024; 39:240-257. [PMID: 38052102 PMCID: PMC10767824 DOI: 10.1093/humrep/dead247] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/14/2023] [Indexed: 12/07/2023] Open
Abstract
STUDY QUESTION Which genetic factors regulate female propensity for giving birth to spontaneous dizygotic (DZ) twins? SUMMARY ANSWER We identified four new loci, GNRH1, FSHR, ZFPM1, and IPO8, in addition to previously identified loci, FSHB and SMAD3. WHAT IS KNOWN ALREADY The propensity to give birth to DZ twins runs in families. Earlier, we reported that FSHB and SMAD3 as associated with DZ twinning and female fertility measures. STUDY DESIGN, SIZE, DURATION We conducted a genome-wide association meta-analysis (GWAMA) of mothers of spontaneous dizygotic (DZ) twins (8265 cases, 264 567 controls) and of independent DZ twin offspring (26 252 cases, 417 433 controls). PARTICIPANTS/MATERIALS, SETTING, METHODS Over 700 000 mothers of DZ twins, twin individuals and singletons from large cohorts in Australia/New Zealand, Europe, and the USA were carefully screened to exclude twins born after use of ARTs. Genetic association analyses by cohort were followed by meta-analysis, phenome wide association studies (PheWAS), in silico and in vivo annotations, and Zebrafish functional validation. MAIN RESULTS AND THE ROLE OF CHANCE This study enlarges the sample size considerably from previous efforts, finding four genome-wide significant loci, including two novel signals and a further two novel genes that are implicated by gene level enrichment analyses. The novel loci, GNRH1 and FSHR, have well-established roles in female reproduction whereas ZFPM1 and IPO8 have not previously been implicated in female fertility. We found significant genetic correlations with multiple aspects of female reproduction and body size as well as evidence for significant selection against DZ twinning during human evolution. The 26 top single nucleotide polymorphisms (SNPs) from our GWAMA in European-origin participants weakly predicted the crude twinning rates in 47 non-European populations (r = 0.23 between risk score and population prevalence, s.e. 0.11, 1-tail P = 0.058) indicating that genome-wide association studies (GWAS) are needed in African and Asian populations to explore the causes of their respectively high and low DZ twinning rates. In vivo functional tests in zebrafish for IPO8 validated its essential role in female, but not male, fertility. In most regions, risk SNPs linked to known expression quantitative trait loci (eQTLs). Top SNPs were associated with in vivo reproductive hormone levels with the top pathways including hormone ligand binding receptors and the ovulation cycle. LARGE SCALE DATA The full DZT GWAS summary statistics will made available after publication through the GWAS catalog (https://www.ebi.ac.uk/gwas/). LIMITATIONS, REASONS FOR CAUTION Our study only included European ancestry cohorts. Inclusion of data from Africa (with the highest twining rate) and Asia (with the lowest rate) would illuminate further the biology of twinning and female fertility. WIDER IMPLICATIONS OF THE FINDINGS About one in 40 babies born in the world is a twin and there is much speculation on why twinning runs in families. We hope our results will inform investigations of ovarian response in new and existing ARTs and the causes of female infertility. STUDY FUNDING/COMPETING INTEREST(S) Support for the Netherlands Twin Register came from the Netherlands Organization for Scientific Research (NWO) and The Netherlands Organization for Health Research and Development (ZonMW) grants, 904-61-193, 480-04-004, 400-05-717, Addiction-31160008, 911-09-032, Biobanking and Biomolecular Resources Research Infrastructure (BBMRI.NL, 184.021.007), Royal Netherlands Academy of Science Professor Award (PAH/6635) to DIB, European Research Council (ERC-230374), Rutgers University Cell and DNA Repository (NIMH U24 MH068457-06), the Avera Institute, Sioux Falls, South Dakota (USA) and the National Institutes of Health (NIH R01 HD042157-01A1) and the Genetic Association Information Network (GAIN) of the Foundation for the National Institutes of Health and Grand Opportunity grants 1RC2 MH089951. The QIMR Berghofer Medical Research Institute (QIMR) study was supported by grants from the National Health and Medical Research Council (NHMRC) of Australia (241944, 339462, 389927, 389875, 389891, 389892, 389938, 443036, 442915, 442981, 496610, 496739, 552485, 552498, 1050208, 1075175). L.Y. is funded by Australian Research Council (Grant number DE200100425). The Minnesota Center for Twin and Family Research (MCTFR) was supported in part by USPHS Grants from the National Institute on Alcohol Abuse and Alcoholism (AA09367 and AA11886) and the National Institute on Drug Abuse (DA05147, DA13240, and DA024417). The Women's Genome Health Study (WGHS) was funded by the National Heart, Lung, and Blood Institute (HL043851 and HL080467) and the National Cancer Institute (CA047988 and UM1CA182913), with support for genotyping provided by Amgen. Data collection in the Finnish Twin Registry has been supported by the Wellcome Trust Sanger Institute, the Broad Institute, ENGAGE-European Network for Genetic and Genomic Epidemiology, FP7-HEALTH-F4-2007, grant agreement number 201413, National Institute of Alcohol Abuse and Alcoholism (grants AA-12502, AA-00145, AA-09203, AA15416, and K02AA018755) and the Academy of Finland (grants 100499, 205585, 118555, 141054, 264146, 308248, 312073 and 336823 to J. Kaprio). TwinsUK is funded by the Wellcome Trust, Medical Research Council, Versus Arthritis, European Union Horizon 2020, Chronic Disease Research Foundation (CDRF), Zoe Ltd and the National Institute for Health Research (NIHR) Clinical Research Network (CRN) and Biomedical Research Centre based at Guy's and St Thomas' NHS Foundation Trust in partnership with King's College London. For NESDA, funding was obtained from the Netherlands Organization for Scientific Research (Geestkracht program grant 10000-1002), the Center for Medical Systems Biology (CSMB, NVVO Genomics), Biobanking and Biomolecular Resources Research Infrastructure (BBMRI-NL), VU University's Institutes for Health and Care Research (EMGO+) and Neuroscience Campus Amsterdam, University Medical Center Groningen, Leiden University Medical Center, National Institutes of Health (NIH, ROI D0042157-01A, MH081802, Grand Opportunity grants 1 RC2 Ml-1089951 and IRC2 MH089995). Part of the genotyping and analyses were funded by the Genetic Association Information Network (GAIN) of the Foundation for the National Institutes of Health. Computing was supported by BiG Grid, the Dutch e-Science Grid, which is financially supported by NWO. Work in the Del Bene lab was supported by the Programme Investissements d'Avenir IHU FOReSIGHT (ANR-18-IAHU-01). C.R. was supported by an EU Horizon 2020 Marie Skłodowska-Curie Action fellowship (H2020-MSCA-IF-2014 #661527). H.S. and K.S. are employees of deCODE Genetics/Amgen. The other authors declare no competing financial interests. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Hamdi Mbarek
- Department of Biological Psychology, Netherlands Twin Register, Vrije Universiteit, Amsterdam, The Netherlands
- Qatar Genome Program, Qatar Foundation, Doha, Qatar
- Amsterdam Reproduction and Development Institute, Amsterdam, The Netherlands
| | - Scott D Gordon
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - David L Duffy
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Nikki Hubers
- Department of Biological Psychology, Netherlands Twin Register, Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Institute, Amsterdam, The Netherlands
| | - Sally Mortlock
- Institute of Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Jeffrey J Beck
- Avera Institute for Human Genetics, Avera McKennan Hospital and University Health Center, Sioux Falls, SD, USA
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, Netherlands Twin Register, Vrije Universiteit, Amsterdam, The Netherlands
| | - René Pool
- Department of Biological Psychology, Netherlands Twin Register, Vrije Universiteit, Amsterdam, The Netherlands
| | - Conor V Dolan
- Department of Biological Psychology, Netherlands Twin Register, Vrije Universiteit, Amsterdam, The Netherlands
| | - Ky’Era V Actkins
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA
| | | | - Jenny Van Dongen
- Department of Biological Psychology, Netherlands Twin Register, Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Institute, Amsterdam, The Netherlands
| | - Erik A Ehli
- Avera Institute for Human Genetics, Avera McKennan Hospital and University Health Center, Sioux Falls, SD, USA
| | - William G Iacono
- Department of Psychology, University of Minnesota, Minneapolis, MN, USA
| | - Matt Mcgue
- Department of Psychology, University of Minnesota, Minneapolis, MN, USA
| | - Daniel I Chasman
- Harvard Medical School, Harvard University, Boston, MA, USA
- Brigham and Women’s Hospital, Boston, MA, USA
| | | | - Samantha L P Schilit
- Harvard Medical School, Harvard University, Boston, MA, USA
- Brigham and Women’s Hospital, Boston, MA, USA
| | - Cynthia C Morton
- Harvard Medical School, Harvard University, Boston, MA, USA
- Brigham and Women’s Hospital, Boston, MA, USA
| | - Guillaume Paré
- Population Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Gonneke Willemsen
- Department of Biological Psychology, Netherlands Twin Register, Vrije Universiteit, Amsterdam, The Netherlands
| | | | | | | | | | | | | | - Eva Krapohl
- Medical Research Council Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
- Statistical Sciences & Innovation, UCB Biosciences GmbH, Monheim, Germany
| | - Robert Plomin
- Medical Research Council Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Patrik K E Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Nancy L Pedersen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Pirro Hysi
- Department of Twin Research & Genetic Epidemiology, King’s College London, London, UK
| | - Massimo Mangino
- Department of Twin Research & Genetic Epidemiology, King’s College London, London, UK
- NIHR Biomedical Research Centre at Guy’s and St Thomas’ Foundation Trust, London, UK
| | - Timothy D Spector
- Department of Twin Research & Genetic Epidemiology, King’s College London, London, UK
| | - Teemu Palviainen
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland
| | - Yuri Milaneschi
- Department of Psychiatry, EMGO Institute for Health and Care Research, Vrije Universiteit, Amsterdam, The Netherlands
| | - Brenda W Penninnx
- Department of Psychiatry, EMGO Institute for Health and Care Research, Vrije Universiteit, Amsterdam, The Netherlands
| | - Adrian I Campos
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Institute of Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Ken K Ong
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - John R B Perry
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Cornelis B Lambalk
- Amsterdam Reproduction and Development Institute, Amsterdam, The Netherlands
- Amsterdam University Medical Centers Location VU Medical Center, Amsterdam, The Netherlands
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland
| | - Ísleifur Ólafsson
- Department of Clinical Biochemistry, National University Hospital of Iceland, Reykjavik, Iceland
| | - Karine Duroure
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Céline Revenu
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | | | - Loic Yengo
- Institute of Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Lea Davis
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA
| | - Eske M Derks
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Sarah E Medland
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | | | | | - Filippo Del Bene
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Bruno Reversade
- Genome Institute of Singapore, Laboratory of Human Genetics & Therapeutics, A*STAR, Singapore, Singapore
- Smart-Health Initiative, BESE, KAUST, Thuwal, Saudi Arabia
| | - Grant W Montgomery
- Institute of Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Dorret I Boomsma
- Department of Biological Psychology, Netherlands Twin Register, Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Institute, Amsterdam, The Netherlands
| | | |
Collapse
|
14
|
Ojala R, Hentilä J, Lietzén MS, Arponen M, Heiskanen MA, Honkala SM, Virtanen H, Koskensalo K, Lautamäki R, Löyttyniemi E, Parkkola R, Heinonen OJ, Malm T, Lahti L, Rinne J, Eskola O, Rajander J, Pietiläinen KH, Kaprio J, Ivaska KK, Hannukainen JC. Bone marrow metabolism is affected by body weight and response to exercise training varies according to anatomical location. Diabetes Obes Metab 2024; 26:251-261. [PMID: 37818602 DOI: 10.1111/dom.15311] [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] [Received: 06/30/2023] [Revised: 09/09/2023] [Accepted: 09/19/2023] [Indexed: 10/12/2023]
Abstract
AIM High body weight is a protective factor against osteoporosis, but obesity also suppresses bone metabolism and whole-body insulin sensitivity. However, the impact of body weight and regular training on bone marrow (BM) glucose metabolism is unclear. We studied the effects of regular exercise training on bone and BM metabolism in monozygotic twin pairs discordant for body weight. METHODS We recruited 12 monozygotic twin pairs (mean ± SD age 40.4 ± 4.5 years; body mass index 32.9 ± 7.6, mean difference between co-twins 7.6 kg/m2 ; eight female pairs). Ten pairs completed the 6-month long training intervention. We measured lumbar vertebral and femoral BM insulin-stimulated glucose uptake (GU) using 18 F-FDG positron emission tomography, lumbar spine bone mineral density and bone turnover markers. RESULTS At baseline, heavier co-twins had higher lumbar vertebral BM GU (p < .001) and lower bone turnover markers (all p < .01) compared with leaner co-twins but there was no significant difference in femoral BM GU, or bone mineral density. Training improved whole-body insulin sensitivity, aerobic capacity (both p < .05) and femoral BM GU (p = .008). The training response in lumbar vertebral BM GU was different between the groups (time × group, p = .02), as GU tended to decrease in heavier co-twins (p = .06) while there was no change in leaner co-twins. CONCLUSIONS In this study, regular exercise training increases femoral BM GU regardless of weight and genetics. Interestingly, lumbar vertebral BM GU is higher in participants with higher body weight, and training counteracts this effect in heavier co-twins even without reduction in weight. These data suggest that BM metabolism is altered by physical activity.
Collapse
Affiliation(s)
- Ronja Ojala
- Turku PET Centre, University of Turku, Turku, Finland
| | | | | | - Milja Arponen
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Marja A Heiskanen
- Turku PET Centre, University of Turku, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | | | | | - Kalle Koskensalo
- Department of Medical Physics, Turku University Hospital, Turku, Finland
| | | | | | - Riitta Parkkola
- Department of Radiology, University of Turku, Turku, Finland
- Department of Radiology, Turku University Hospital, Turku, Finland
| | - Olli J Heinonen
- Paavo Nurmi Centre, Department of Health and Physical Activity, University of Turku, Turku, Finland
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Leo Lahti
- Department of Computing, University of Turku, Turku, Finland
| | - Juha Rinne
- Turku PET Centre, University of Turku, Turku, Finland
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Olli Eskola
- Radiopharmaceutical Chemistry Laboratory, Turku PET Centre, University of Turku, Turku, Finland
| | - Johan Rajander
- Turku PET Centre, Accelerator Laboratory, Åbo Akademi University, Turku, Finland
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Healthy Weight Hub, Abdominal Center, Endocrinology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland FIMM, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Kaisa K Ivaska
- Institute of Biomedicine, University of Turku, Turku, Finland
| | | |
Collapse
|
15
|
Duncan GE, Avery A, Maamar MB, Nilsson EE, Beck D, Skinner MK. Epigenome-wide association study of systemic effects of obesity susceptibility in human twins. Epigenetics 2023; 18:2268834. [PMID: 37871278 PMCID: PMC10595392 DOI: 10.1080/15592294.2023.2268834] [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: 03/20/2023] [Accepted: 10/01/2023] [Indexed: 10/25/2023] Open
Abstract
The current study was designed to use an epigenome-wide association approach (EWAS) to identify potential systemic DNA methylation alterations that are associated with obesity using 22 discordant twin pairs. Buccal cells (from a cheek swab) were used as a non-obesity relevant purified marker cell for the epigenetic analysis. Analysis of differential DNA methylation regions (DMRs) was used to identify epigenetic associations with metabolic and dietary measures related to obesity with discordant twins. An edgeR analysis provided a DMR signature with p < 1e-04, but statistical significance was reduced due to low sample size and known multiple origins of obesity. A weighted gene coexpression network analysis (WGCNA) was performed and identified modules (p < 0.005) of epigenetic sites that correlated with different metabolic and dietary measures. The DMR and WGCNA epigenetic sites were near genes (e.g., CIDEC, SPP1, ZFPG9, and POMC) with previously identified obesity associated pathways (e.g., metabolism, cholesterol, and fat digestion). Observations demonstrate the feasibility of identifying systemic epigenetic biomarkers for obesity, which can be further investigated for clinical relevance in future research with larger sample sizes. The availability of a systemic epigenetic biomarker for obesity susceptibility may facilitate preventative medicine and clinical management of the disease early in life.
Collapse
Affiliation(s)
- Glen E. Duncan
- Department of Nutrition and Exercise Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Ally Avery
- Department of Nutrition and Exercise Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Millissia Ben Maamar
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA, USA
| | - Eric E. Nilsson
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA, USA
| | - Daniel Beck
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA, USA
| | - Michael K. Skinner
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA, USA
| |
Collapse
|
16
|
Sehovic E, Zellers SM, Youssef MK, Heikkinen A, Kaprio J, Ollikainen M. DNA methylation sites in early adulthood characterised by pubertal timing and development: a twin study. Clin Epigenetics 2023; 15:181. [PMID: 37950287 PMCID: PMC10638786 DOI: 10.1186/s13148-023-01594-7] [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: 07/17/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Puberty is a highly heritable and variable trait, with environmental factors having a role in its eventual timing and development. Early and late pubertal onset are both associated with various diseases developing later in life, and epigenetic characterisation of pubertal timing and development could lead to important insights. Blood DNA methylation, reacting to both genotype and environment, has been associated with puberty; however, such studies are relatively scarce. We investigated peripheral blood DNA methylation profiles (using Illumina 450 K and EPIC platforms) of 1539 young adult Finnish twins associated with pubertal development scale (PDS) at ages 12 and 14 as well as pubertal age (PA). RESULTS Fixed effect meta-analysis of the two platforms on 347,521 CpGs in common identified 58 CpG sites associated (p < 1 × 10-5) with either PDS or PA. All four CpGs associated with PA and 45 CpGs associated with PDS were sex-specific. Thirteen CpGs had a high heritability (h2: 0.51-0.98), while one CpG site (mapped to GET4) had a high shared environmental component accounting for 68% of the overall variance in methylation at the site. Utilising twin discordance analysis, we found 6 CpG sites (5 associated with PDS and 1 with PA) that had an environmentally driven association with puberty. Furthermore, genes with PDS- or PA-associated CpGs were consistently linked to various developmental processes and diseases such as breast, prostate and ovarian cancer, while methylation quantitative trait loci of associated CpG sites were enriched in immune pathways developing during puberty. CONCLUSIONS By identifying puberty-associated DNA methylation sites and examining the effects of sex, environment and genetics, we shed light on the intricate interplay between environment and genetics in the context of puberty. Through our comprehensive analysis, we not only deepen the understanding of the significance of both genetic and environmental factors in the complex processes of puberty and its timing, but also gain insights into potential links with disease risks.
Collapse
Affiliation(s)
- Emir Sehovic
- Department of Life Sciences and Systems Biology, University of Turin, 10100, Turin, Italy
- Cancer Genomics Lab, Fondazione Edo ed Elvo Tempia, 13900, Biella, Italy
| | - Stephanie M Zellers
- Institute for Molecular Medicine Finland, University of Helsinki, 00290, Helsinki, Finland
| | - Markus K Youssef
- Laboratory for Topology and Neuroscience, Brain Mind Institute, EPFL, 1015, Lausanne, Switzerland
| | - Aino Heikkinen
- Institute for Molecular Medicine Finland, University of Helsinki, 00290, Helsinki, Finland
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland, University of Helsinki, 00290, Helsinki, Finland
| | - Miina Ollikainen
- Institute for Molecular Medicine Finland, University of Helsinki, 00290, Helsinki, Finland.
- Minerva Foundation Institute for Medical Research, 00290, Helsinki, Finland.
| |
Collapse
|
17
|
Hirvelä L, Keski-Rahkonen A, Sipilä PN. Associations of broad eating disorder symptoms with later alcohol problems in Finnish adult twins: A nationwide 10-year follow-up. Int J Eat Disord 2023; 56:1854-1865. [PMID: 37353472 DOI: 10.1002/eat.24013] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 06/25/2023]
Abstract
OBJECTIVE Eating disorders are associated with subsequent alcohol problems, but it is not known whether this association also extends to broader eating disorder symptoms not captured by clinical diagnoses. We assessed the longitudinal association of broad eating disorder symptoms with alcohol problems in a nationwide twin sample (FinnTwin16). METHODS Finnish women (N = 1905) and men (N = 1449) self-reported their eating disorder symptoms using the Eating Disorder Inventory-2 Bulimia, Drive for Thinness, and Body Dissatisfaction subscales at the mean age of 24.4 years in 2000-2003. A subsample of participants also completed items on drive for muscularity, height dissatisfaction, and muscle-enhancing supplement use. Alcohol problems were assessed 10 years later at the age of 34.1 in 2010-2012 with the Rutgers Alcohol Problem Index. RESULTS Eating disorder symptoms were associated with later alcohol problems (odds ratio per point increase 1.02-1.18). Bulimia showed stronger associations among men than women (p for interaction .012). Drive for muscularity and height dissatisfaction were also associated with later alcohol problems, but supplement use was not. When accounting for baseline alcohol problems, only Bulimia (among women and men) and Drive for Thinness (among men) were significantly associated with later alcohol problems. Bulimia was also significantly associated with later alcohol problems in within-twin-pair analyses among dizygotic twins, but not among monozygotic twins. DISCUSSION In a longitudinal setting, eating disorder symptoms were associated with later alcohol problems. Bulimic symptoms were a stronger risk factor for men than women. These associations may be attributable to baseline alcohol problems, childhood environment and genetic liability. PUBLIC SIGNIFICANCE This study found that both young adult women and men with broad eating disorder symptoms are at a higher risk of alcohol-related problems than those without such symptoms. Men with bulimic symptoms were at a particularly high risk. These findings emphasize the need for better prevention and treatment of disordered eating, body image concerns and alcohol problems for both young adult women and men.
Collapse
Affiliation(s)
- Leon Hirvelä
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | | | - Pyry N Sipilä
- Department of Public Health, University of Helsinki, Helsinki, Finland
| |
Collapse
|
18
|
Jersin RÅ, Sri Priyanka Tallapragada D, Skartveit L, Bjune MS, Muniandy M, Lee-Ødegård S, Heinonen S, Alvarez M, Birkeland KI, André Drevon C, Pajukanta P, McCann A, Pietiläinen KH, Claussnitzer M, Mellgren G, Dankel SN. Impaired Adipocyte SLC7A10 Promotes Lipid Storage in Association With Insulin Resistance and Altered BCAA Metabolism. J Clin Endocrinol Metab 2023; 108:2217-2229. [PMID: 36916878 PMCID: PMC10438883 DOI: 10.1210/clinem/dgad148] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 02/23/2023] [Accepted: 03/07/2023] [Indexed: 03/16/2023]
Abstract
CONTEXT The neutral amino acid transporter SLC7A10/ASC-1 is an adipocyte-expressed gene with reduced expression in insulin resistance and obesity. Inhibition of SLC7A10 in adipocytes was shown to increase lipid accumulation despite decreasing insulin-stimulated uptake of glucose, a key substrate for de novo lipogenesis. These data imply that alternative lipogenic substrates to glucose fuel continued lipid accumulation during insulin resistance in obesity. OBJECTIVE We examined whether increased lipid accumulation during insulin resistance in adipocytes may involve alter flux of lipogenic amino acids dependent on SLC7A10 expression and activity, and whether this is reflected by extracellular and circulating concentrations of marker metabolites. METHODS In adipocyte cultures with impaired SLC7A10, we performed RNA sequencing and relevant functional assays. By targeted metabolite analyses (GC-MS/MS), flux of all amino acids and selected metabolites were measured in human and mouse adipose cultures. Additionally, SLC7A10 mRNA levels in human subcutaneous adipose tissue (SAT) were correlated to candidate metabolites and adiposity phenotypes in 2 independent cohorts. RESULTS SLC7A10 impairment altered expression of genes related to metabolic processes, including branched-chain amino acid (BCAA) catabolism, lipogenesis, and glyceroneogenesis. In 3T3-L1 adipocytes, SLC7A10 inhibition increased fatty acid uptake and cellular content of glycerol and cholesterol. SLC7A10 impairment in SAT cultures altered uptake of aspartate and glutamate, and increased net uptake of BCAAs, while increasing the net release of the valine catabolite 3- hydroxyisobutyrate (3-HIB). In human cohorts, SLC7A10 mRNA correlated inversely with total fat mass, circulating triacylglycerols, BCAAs, and 3-HIB. CONCLUSION Reduced SLC7A10 activity strongly affects flux of BCAAs in adipocytes, which may fuel continued lipogenesis during insulin resistance, and be reflected in increased circulating levels of the valine-derived catabolite 3-HIB.
Collapse
Affiliation(s)
- Regine Å Jersin
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Divya Sri Priyanka Tallapragada
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Linn Skartveit
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Mona S Bjune
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Maheswary Muniandy
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland
| | - Sindre Lee-Ødegård
- Department of Transplantation Medicine, The University of Oslo, Institute of Clinical Medicine, and Oslo University Hospital, N-0372 Oslo, Norway
| | - Sini Heinonen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland
| | - Marcus Alvarez
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Kåre Inge Birkeland
- Department of Transplantation Medicine, The University of Oslo, Institute of Clinical Medicine, and Oslo University Hospital, N-0372 Oslo, Norway
| | - Christian André Drevon
- Department of Nutrition, The University of Oslo, Institute of Basic Medical Sciences, N-0372 Oslo, Norway
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA 90095, USA
- Institute for Precision Health, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Adrian McCann
- Bevital A/S, Laboratoriebygget, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland
- Obesity Center, Endocrinology, Abdominal Center, Helsinki University Hospital and University of Helsinki, FIN-00014 Helsinki, Finland
| | - Melina Claussnitzer
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Gunnar Mellgren
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, N-5021 Bergen, Norway
| | - Simon N Dankel
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, N-5021 Bergen, Norway
| |
Collapse
|
19
|
Garske KM, Kar A, Comenho C, Balliu B, Pan DZ, Bhagat YV, Rosenberg G, Koka A, Das SS, Miao Z, Sinsheimer JS, Kaprio J, Pietiläinen KH, Pajukanta P. Increased body mass index is linked to systemic inflammation through altered chromatin co-accessibility in human preadipocytes. Nat Commun 2023; 14:4214. [PMID: 37452040 PMCID: PMC10349101 DOI: 10.1038/s41467-023-39919-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 07/04/2023] [Indexed: 07/18/2023] Open
Abstract
Obesity-induced adipose tissue dysfunction can cause low-grade inflammation and downstream obesity comorbidities. Although preadipocytes may contribute to this pro-inflammatory environment, the underlying mechanisms are unclear. We used human primary preadipocytes from body mass index (BMI) -discordant monozygotic (MZ) twin pairs to generate epigenetic (ATAC-sequence) and transcriptomic (RNA-sequence) data for testing whether increased BMI alters the subnuclear compartmentalization of open chromatin in the twins' preadipocytes, causing downstream inflammation. Here we show that the co-accessibility of open chromatin, i.e. compartmentalization of chromatin activity, is altered in the higher vs lower BMI MZ siblings for a large subset ( ~ 88.5 Mb) of the active subnuclear compartments. Using the UK Biobank we show that variants within these regions contribute to systemic inflammation through interactions with BMI on C-reactive protein. In summary, open chromatin co-accessibility in human preadipocytes is disrupted among the higher BMI siblings, suggesting a mechanism how obesity may lead to inflammation via gene-environment interactions.
Collapse
Affiliation(s)
- Kristina M Garske
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Asha Kar
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Caroline Comenho
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Brunilda Balliu
- Department of Computational Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - David Z Pan
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, 90095, USA
| | - Yash V Bhagat
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Gregory Rosenberg
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Amogha Koka
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Sankha Subhra Das
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Zong Miao
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, 90095, USA
| | - Janet S Sinsheimer
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
- Department of Computational Medicine, UCLA, Los Angeles, CA, 90095, USA
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, 90095, USA
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, 00014, Finland
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, 00014, Finland
- Obesity Center, Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, 00014, Finland
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA.
- Department of Computational Medicine, UCLA, Los Angeles, CA, 90095, USA.
- Institute for Precision Heath, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA.
| |
Collapse
|
20
|
Gonggrijp BMA, Silventoinen K, Dolan CV, Boomsma DI, Kaprio J, Willemsen G. The mechanism of assortative mating for educational attainment: a study of Finnish and Dutch twins and their spouses. Front Genet 2023; 14:1150697. [PMID: 37396041 PMCID: PMC10311485 DOI: 10.3389/fgene.2023.1150697] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 05/31/2023] [Indexed: 07/04/2023] Open
Abstract
Introduction: Assortative mating refers describes a phenomenon in which individuals with similar phenotypic traits are more likely to mate and reproduce with each other; i.e. assortative mating occurs when individuals choose partners based on their similarity or dissimilarity in certain traits.to patterns of non-random mating of spouses leading to phenotypic resemblance. There are various theories about the its underlying mechanisms, which have different genetic consequences. Methods: We analyzed examined two possible mechanisms underlying assortative mating - phenotypic assortment and social homogamy - for educational attainment in two countries utilizing data of mono- and dizygotic twins and their spouses (1,451 Finnish and 1,616 Dutch twin-spouse pairs). Results: The spousal correlations were 0.51 in Finland and 0.45 in the Netherlands, to which phenotypic assortment contributed 0.35 and 0.30, and social homogamy 0.16 and 0.15, respectively. Conclusion: Both social homogamy and phenotypic assortment are important processes in spouse selection in Finland and the Netherlands. In both countries, phenotypic assortment contributes to a greater degree to the similarity of spouses than social homogamy.
Collapse
Affiliation(s)
- Bodine M. A. Gonggrijp
- Netherlands Institute for the Study of Crime and Law Enforcement (NSCR), Amsterdam, Netherlands
- Department of Biological Psychology, Faculty of Behavioural and Movement Sciences, VU Amsterdam, Amsterdam, Netherlands, Netherlands
| | - Karri Silventoinen
- Faculty of Social Sciences, University of Helsinki, Helsinki, Uusimaa, Finland
- Department of Public Health, Faculty of Medicine, University of Helsinki, Helsinki, Uusimaa, Finland
| | - Conor V. Dolan
- Department of Biological Psychology, Faculty of Behavioural and Movement Sciences, VU Amsterdam, Amsterdam, Netherlands, Netherlands
| | - Dorret I. Boomsma
- Department of Biological Psychology, Faculty of Behavioural and Movement Sciences, VU Amsterdam, Amsterdam, Netherlands, Netherlands
- Amsterdam Public Health Research Institute, VU Medical Center, Amsterdam, Netherlands
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Uusimaa, Finland
| | - Gonneke Willemsen
- Department of Biological Psychology, Faculty of Behavioural and Movement Sciences, VU Amsterdam, Amsterdam, Netherlands, Netherlands
- Amsterdam Public Health Research Institute, VU Medical Center, Amsterdam, Netherlands
| |
Collapse
|
21
|
Sipilä PN, Keski-Rahkonen A, Lindbohm JV, Rose RJ, Kaprio J. Paternal and Maternal Problem Drinking and Lifetime Problem Drinking of Their Adult Children. Twin Res Hum Genet 2023; 26:152-163. [PMID: 37092738 PMCID: PMC11000696 DOI: 10.1017/thg.2023.12] [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] [Indexed: 04/25/2023]
Abstract
Parents' alcohol use is associated with alcohol use of their adolescent offspring, but does this association extend to the adulthood of the offspring? We examined associations of paternal and maternal problem drinking with lifetime problem drinking of their adult offspring prospectively assessed in a population-based Finnish twin-family cohort (FinnTwin16). Problem drinking (Malmö-modified Michigan Alcoholism Screening Test) was self-reported separately by mothers and fathers when their children were 16. The children reported on an extended lifetime version of the same measure during their mid-twenties (21-28 years) and mid-thirties (31-37 years). 1235 sons and 1461 daughters in mid-twenties and 991 sons and 1278 daughters in mid-thirties had complete data. Correlations between fathers' and their adult children's problem drinking ranged from .12 to .18. For mothers and their adult children, these correlations ranged from .09 to .14. In multivariate models, adjustment for potential confounders had little effect on the observed associations. In this study, parental problem drinking was modestly associated with lifetime problem drinking of their adult children. This association could be detected even when the children had reached the fourth decade of life.
Collapse
Affiliation(s)
- Pyry N Sipilä
- Clinicum, Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Anna Keski-Rahkonen
- Clinicum, Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Joni V Lindbohm
- Clinicum, Department of Public Health, University of Helsinki, Helsinki, Finland
- Department of Epidemiology and Public Health, University College London, London, United Kingdom
| | - Richard J Rose
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana, USA
| | - Jaakko Kaprio
- Clinicum, Department of Public Health, University of Helsinki, Helsinki, Finland
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| |
Collapse
|
22
|
Marttila S, Tamminen H, Rajić S, Mishra PP, Lehtimäki T, Raitakari O, Kähönen M, Kananen L, Jylhävä J, Hägg S, Delerue T, Peters A, Waldenberger M, Kleber ME, März W, Luoto R, Raitanen J, Sillanpää E, Laakkonen EK, Heikkinen A, Ollikainen M, Raitoharju E. Methylation status of VTRNA2-1/ nc886 is stable across populations, monozygotic twin pairs and in majority of tissues. Epigenomics 2022; 14:1105-1124. [PMID: 36200237 DOI: 10.2217/epi-2022-0228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aims & methods: The aim of this study was to characterize the methylation level of a polymorphically imprinted gene, VTRNA2-1/nc886, in human populations and somatic tissues.48 datasets, consisting of more than 30 tissues and >30,000 individuals, were used. Results: nc886 methylation status is associated with twin status and ethnic background, but the variation between populations is limited. Monozygotic twin pairs present concordant methylation, whereas ∼30% of dizygotic twin pairs present discordant methylation in the nc886 locus. The methylation levels of nc886 are uniform across somatic tissues, except in cerebellum and skeletal muscle. Conclusion: The nc886 imprint may be established in the oocyte, and, after implantation, the methylation status is stable, excluding a few specific tissues.
Collapse
Affiliation(s)
- Saara Marttila
- Molecular Epidemiology, Faculty of Medicine & Health Technology, Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland.,Gerontology Research Center, Tampere University, Tampere, 33014, Finland
| | - Hely Tamminen
- Molecular Epidemiology, Faculty of Medicine & Health Technology, Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland
| | - Sonja Rajić
- Molecular Epidemiology, Faculty of Medicine & Health Technology, Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland
| | - Pashupati P Mishra
- Department of Clinical Chemistry, Faculty of Medicine & Health Technology, Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland.,Finnish Cardiovascular Research Center Tampere, Faculty of Medicine & Health Technology, Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland.,Fimlab Laboratories, Arvo Ylpön katu 4, Tampere, 33520, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Faculty of Medicine & Health Technology, Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland.,Finnish Cardiovascular Research Center Tampere, Faculty of Medicine & Health Technology, Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland.,Fimlab Laboratories, Arvo Ylpön katu 4, Tampere, 33520, Finland
| | - Olli Raitakari
- Centre for Population Health Research, University of Turku & Turku University Hospital, Turku, 20014, Finland.,Research Centre of Applied & Preventive Cardiovascular Medicine, University of Turku, Turku, 20014, Finland.,Department of Clinical Physiology & Nuclear Medicine, Turku University Hospital, Turku, 20014, Finland
| | - Mika Kähönen
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine & Health Technology, Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland.,Department of Clinical Physiology, Tampere University Hospital, Tampere, 33521, Finland
| | - Laura Kananen
- Faculty of Medicine & Health Technology, & Gerontology Research Center, Tampere University, Arvo Ylpön katu 34, Tampere, 33520,Finland.,Department of Medical Epidemiology & Biostatistics, Karolinska Institutet, Stockholm, 171 77, Sweden.,Faculty of Social Sciences (Health Sciences), & Gerontology Research Center, Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland
| | - Juulia Jylhävä
- Department of Medical Epidemiology & Biostatistics, Karolinska Institutet, Stockholm, 171 77, Sweden.,Faculty of Social Sciences (Health Sciences), & Gerontology Research Center, Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland
| | - Sara Hägg
- Department of Medical Epidemiology & Biostatistics, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Thomas Delerue
- Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, D-85764,, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, D-85764, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Melanie Waldenberger
- Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria, D-85764,, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Marcus E Kleber
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany.,SYNLAB MVZ Humangenetik Mannheim, Mannheim, Germany
| | - Winfried März
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany.,Competence Cluster for Nutrition & Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Jena, 07743, Germany.,SYNLAB Academy, SYNLAB Holding Deutschland GmbH, Augsburg, 86156, Germany.,Clinical Institute of Medical & Chemical Laboratory Diagnostics, Medical University of Graz, Graz, 8010, Austria
| | - Riitta Luoto
- The Social Insurance Institute of Finland (Kela), Helsinki, 00250, Finland.,The UKK Institute for Health Promotion Research, Kaupinpuistonkatu 1, Tampere, 33500, Finland
| | - Jani Raitanen
- The UKK Institute for Health Promotion Research, Kaupinpuistonkatu 1, Tampere, 33500, Finland.,Faculty of Social Sciences (Health Sciences), Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland
| | - Elina Sillanpää
- Gerontology Research Center & Faculty of Sport & Health Sciences, University of Jyväskylä, Jyväskylä, 40014, Finland.,Institute for Molecular Medicine Finland, FIMM, HiLIFE, University of Helsinki, Helsinki, 00014, Finland
| | - Eija K Laakkonen
- Gerontology Research Center & Faculty of Sport & Health Sciences, University of Jyväskylä, Jyväskylä, 40014, Finland
| | - Aino Heikkinen
- Institute for Molecular Medicine Finland, FIMM, HiLIFE, University of Helsinki, Helsinki, 00014, Finland
| | - Miina Ollikainen
- Institute for Molecular Medicine Finland, FIMM, HiLIFE, University of Helsinki, Helsinki, 00014, Finland
| | - Emma Raitoharju
- Molecular Epidemiology, Faculty of Medicine & Health Technology, Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland.,Finnish Cardiovascular Research Center Tampere, Faculty of Medicine & Health Technology, Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland
| |
Collapse
|
23
|
Lundgren S, Kuitunen S, Pietiläinen KH, Hurme M, Kähönen M, Männistö S, Perola M, Lehtimäki T, Raitakari O, Kaprio J, Ollikainen M. BMI is positively associated with accelerated epigenetic aging in twin pairs discordant for body mass index. J Intern Med 2022; 292:627-640. [PMID: 35699258 PMCID: PMC9540898 DOI: 10.1111/joim.13528] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Obesity is a heritable complex phenotype that can increase the risk of age-related outcomes. Biological age can be estimated from DNA methylation (DNAm) using various "epigenetic clocks." Previous work suggests individuals with elevated weight also display accelerated aging, but results vary by epigenetic clock and population. Here, we utilize the new epigenetic clock GrimAge, which closely correlates with mortality. OBJECTIVES We aimed to assess the cross-sectional association of body mass index (BMI) with age acceleration in twins to limit confounding by genetics and shared environment. METHODS AND RESULTS Participants were from the Finnish Twin Cohort (FTC; n = 1424), including monozygotic (MZ) and dizygotic (DZ) twin pairs, and DNAm was measured using the Illumina 450K array. Multivariate linear mixed effects models including MZ and DZ twins showed an accelerated epigenetic age of 1.02 months (p-value = 6.1 × 10-12 ) per one-unit BMI increase. Additionally, heavier twins in a BMI-discordant MZ twin pair (ΔBMI >3 kg/m2 ) had an epigenetic age 5.2 months older than their lighter cotwin (p-value = 0.0074). We also found a positive association between log (homeostatic model assessment of insulin resistance) and age acceleration, confirmed by a meta-analysis of the FTC and two other Finnish cohorts (overall effect = 0.45 years, p-value = 4.1 × 10-25 ) from adjusted models. CONCLUSION We identified significant associations of BMI and insulin resistance with age acceleration based on GrimAge, which were not due to genetic effects on BMI and aging. Overall, these results support a role of BMI in aging, potentially in part due to the effects of insulin resistance.
Collapse
Affiliation(s)
- Sara Lundgren
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Sara Kuitunen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Obesity Center, Endocrinology, Abdominal Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Mikko Hurme
- Department of Microbiology and Immunology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland.,Finnish Cardiovascular Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Satu Männistö
- Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Markus Perola
- Finnish Institute for Health and Welfare, Helsinki, Finland.,Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Olli Raitakari
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland.,Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland.,Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Miina Ollikainen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
24
|
Pascale A, Stephenson M, Barr P, Latvala A, Aaltonen S, Piirtola M, Viken R, Rose RJ, Kaprio J, Maes H, Dick DM, Salvatore JE. Exploring the relationships between adolescent alcohol misuse and later life health outcomes. Alcohol Clin Exp Res 2022; 46:1753-1765. [PMID: 36115067 PMCID: PMC9509441 DOI: 10.1111/acer.14917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 07/18/2022] [Accepted: 07/25/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND We sought to clarify the impact of adolescent alcohol misuse on adult physical health and subjective well-being. To do so, we investigated both the direct associations between adolescent alcohol misuse and early midlife physical health and life satisfaction and the indirect effects on these outcomes attributable to subsequent alcohol problems. METHOD The sample included 2733 twin pairs (32% monozygotic; 52% female) from the FinnTwin16 study. Adolescent alcohol misuse was a composite of frequency of drunkenness, frequency of alcohol use, and alcohol problems at ages 16, 17, and 18.5. The early midlife outcomes included somatic symptoms, self-rated health, and life satisfaction at age 34. The mediators examined as part of the indirect effect analyses included alcohol problems from the Rutgers Alcohol Problem Index at ages 24 and 34. Serial mediation and co-twin comparison models were applied and included covariates from adolescence and early midlife. RESULTS There were weak direct associations between adolescent alcohol misuse and early midlife physical health and life satisfaction. However, there was stronger evidence for indirect effects, whereby young adult and early midlife alcohol problems serially mediated the relationship between adolescent alcohol misuse and early midlife somatic symptoms (β = 0.03, 95% CI [0.03, 0.04]), self-rated health (β = -0.02, 95% CI [-0.03, -0.01]), and life satisfaction (β = -0.03, CI [-0.04, -0.02]). These serial mediation effects were robust in co-twin comparison analyses. CONCLUSIONS These results provide evidence that alcohol problems are a primary driver linking adolescent alcohol misuse and poor health outcomes across the lifespan.
Collapse
Affiliation(s)
- Angela Pascale
- Department of PsychologyVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Mallory Stephenson
- Department of PsychologyVirginia Commonwealth UniversityRichmondVirginiaUSA
- Virginia Institute for Psychiatric and Behavioral GeneticsRichmondVirginiaUSA
| | - Peter Barr
- Department of Psychiatry and Behavioral SciencesSUNY Downstate Health Sciences UniversityBrooklynNew YorkUSA
| | - Antti Latvala
- Institute of Criminology and Legal PolicyUniversity of HelsinkiHelsinkiFinland
| | - Sari Aaltonen
- Institute for Molecular Medicine Finland (FIMM)University of HelsinkiHelsinkiFinland
| | - Maarit Piirtola
- Institute for Molecular Medicine Finland (FIMM)University of HelsinkiHelsinkiFinland
- UKK Institute for Health Promotion ResearchTampereFinland
| | - Richard Viken
- Department of Psychological and Brain SciencesIndiana UniversityBloomingtonIndianaUSA
| | - Richard J. Rose
- Department of Psychological and Brain SciencesIndiana UniversityBloomingtonIndianaUSA
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland (FIMM)University of HelsinkiHelsinkiFinland
| | - Hermine Maes
- Virginia Institute for Psychiatric and Behavioral GeneticsRichmondVirginiaUSA
| | - Danielle M. Dick
- Department of PsychologyVirginia Commonwealth UniversityRichmondVirginiaUSA
- Department of Human and Molecular GeneticsVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Jessica E. Salvatore
- Department of PsychiatryRutgers Robert Wood Johnson School of MedicinePiscatawayNew JerseyUSA
| |
Collapse
|
25
|
Kujala UM, Leskinen T, Rottensteiner M, Aaltonen S, Ala-Korpela M, Waller K, Kaprio J. Physical activity and health: Findings from Finnish monozygotic twin pairs discordant for physical activity. Scand J Med Sci Sports 2022; 32:1316-1323. [PMID: 35770444 PMCID: PMC9378553 DOI: 10.1111/sms.14205] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 05/21/2022] [Accepted: 06/15/2022] [Indexed: 11/28/2022]
Abstract
Genetic and early environmental differences including early health habits associate with future health. To provide insight on the causal nature of these associations, monozygotic (MZ) twin pairs discordant for health habits provide an interesting natural experiment. Twin pairs discordant for leisure‐time physical activity (LTPA) in early adult life is thus a powerful study design to investigate the associations between long‐term LTPA and indicators of health and wellbeing. We have identified 17 LTPA discordant twin pairs from two Finnish twin cohorts and summarize key findings of these studies in this paper. The carefully characterized rare long‐term LTPA discordant MZ twin pairs have participated in multi‐dimensional clinical examinations. Key findings highlight that compared with less active twins in such MZ twin pairs, the twins with higher long‐term LTPA have higher physical fitness, reduced body fat, reduced visceral fat, reduced liver fat, increased lumen diameters of conduit arteries to the lower limbs, increased bone mineral density in loaded bone areas, and an increased number of large high‐density lipoprotein particles. The findings increase our understanding on the possible site‐specific and system‐level effects of long‐term LTPA.
Collapse
Affiliation(s)
- Urho M Kujala
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Tuija Leskinen
- Department of Public Health, University of Turku and Turku University Hospital, Turku, Finland
| | - Mirva Rottensteiner
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Sari Aaltonen
- Institute for Molecular Medicine FIMM, University of Helsinki, Helsinki, Finland
| | - Mika Ala-Korpela
- Systems Epidemiology, Faculty of Medicine, University of Oulu & Biocenter Oulu, Oulu, Finland.,Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland.,NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Katja Waller
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Jaakko Kaprio
- Institute for Molecular Medicine FIMM, University of Helsinki, Helsinki, Finland
| |
Collapse
|
26
|
Koskinen SM, Ahveninen J, Kujala T, Kaprio J, O'Donnell BF, Osipova D, Viken RJ, Näätänen R, Rose RJ. Association of lifetime major depressive disorder with enhanced attentional sensitivity measured with P3 response in young adult twins. Biol Psychol 2022; 171:108345. [PMID: 35525377 DOI: 10.1016/j.biopsycho.2022.108345] [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: 05/03/2021] [Revised: 04/29/2022] [Accepted: 05/01/2022] [Indexed: 11/02/2022]
Abstract
Major depression is associated with alterations in the auditory P3 event-related potential (ERP). However, the persistence of these abnormalities after recovery from depressive episodes, especially in young adults, is not well known. Furthermore, the potential influence of substance use on this association is poorly understood. Young adult twin pairs (N=177) from the longitudinal FinnTwin16 study were studied with a psychiatric interview, and P3a and P3b ERPs elicited by task-irrelevant novel sounds and targets, respectively. Dyadic linear mixed-effect models were used to distinguish the effects of lifetime major depressive disorder from familial factors and effects of alcohol problem drinking and tobacco smoking. P3a amplitude was significantly increased and P3b latency decreased, in individuals with a history of lifetime major depression, when controlling the fixed effects of alcohol abuse, tobacco, gender, twins' birth order, and zygosity. These results suggest that past lifetime major depressive disorder may be associated with enhanced attentional sensitivity.
Collapse
Affiliation(s)
- Sini M Koskinen
- Cognitive Brain Research Unit, Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, POB 21, FI-00014, Helsinki, Finland.
| | - Jyrki Ahveninen
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, 149 13th St, Charlestown, Massachusetts 02129, USA; Department of Radiology, Harvard Medical School, 25 Shattuck Street, Boston, Massachusetts 02115, USA.
| | - Teija Kujala
- Cognitive Brain Research Unit, Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, POB 21, FI-00014, Helsinki, Finland.
| | - Jaakko Kaprio
- Department of Public Health & Institute for Molecular Medicine, University of Helsinki, POB 4, FI-00014, Helsinki, Finland.
| | - Brian F O'Donnell
- Department of Psychological & Brain Sciences, Indiana University, 1101 E 10th St, Bloomington, Indiana 47405, USA.
| | - Daria Osipova
- Cognitive Brain Research Unit, Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, POB 21, FI-00014, Helsinki, Finland.
| | - Richard J Viken
- Department of Psychological & Brain Sciences, Indiana University, 1101 E 10th St, Bloomington, Indiana 47405, USA.
| | - Risto Näätänen
- Cognitive Brain Research Unit, Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, POB 21, FI-00014, Helsinki, Finland.
| | - Richard J Rose
- Department of Psychological & Brain Sciences, Indiana University, 1101 E 10th St, Bloomington, Indiana 47405, USA.
| |
Collapse
|
27
|
Silventoinen K, Jelenkovic A, Palviainen T, Dunkel L, Kaprio J. The Association Between Puberty Timing and Body Mass Index in a Longitudinal Setting: The Contribution of Genetic Factors. Behav Genet 2022; 52:186-194. [PMID: 35381915 PMCID: PMC9135891 DOI: 10.1007/s10519-022-10100-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 03/17/2022] [Indexed: 12/11/2022]
Abstract
We analyzed the contribution of genetic factors on the association between puberty timing and body mass index (BMI) using longitudinal data and two approaches: (i) genetic twin design and (ii) polygenic scores (PGS) of obesity indices. Our data were derived from Finnish cohorts: 9080 twins had information on puberty timing and BMI and 2468 twins also had genetic data. Early puberty timing was moderately associated with higher BMI in childhood in both boys and girls; in adulthood these correlations were weaker and largely disappeared after adjusting for childhood BMI. The largest proportion of these correlations was attributable to genetic factors. The higher PGSs of BMI and waist circumference were associated with earlier timing of puberty in girls, whereas weaker associations were found in boys. Early puberty is not an independent risk factor for adult obesity but rather reflects the association between puberty timing and childhood BMI contributed by genetic predisposition.
Collapse
Affiliation(s)
- Karri Silventoinen
- Population Research Unit, Faculty of Social Sciences, University of Helsinki, P.O. Box 18, 00014, Helsinki, Finland.
- Department of Public Health, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Aline Jelenkovic
- Department of Public Health, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - Teemu Palviainen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Leo Dunkel
- Barts & the London Medical School, William Harvey Research Institute, London, UK
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
28
|
Hirvelä L, Sipilä PN, Keski-Rahkonen A. Relationship between sensation seeking, alcohol problems and bulimic symptoms: a community-based, longitudinal study. Eat Weight Disord 2022; 27:589-595. [PMID: 33900563 PMCID: PMC8933307 DOI: 10.1007/s40519-021-01193-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 04/10/2021] [Indexed: 10/31/2022] Open
Abstract
PURPOSE The association of bulimic symptoms with sensation seeking is uncertain; however, both behaviors have been linked to alcohol problems. We assessed in a longitudinal, community-based setting whether sensation seeking in adolescence is associated with bulimic symptoms in early adulthood, also accounting for alcohol problems. METHODS Finnish men (N = 2000) and women (N = 2467) born between 1974-1979 completed Zuckerman's sensation seeking scale (SSS) at age 18. Alcohol problems (Malmö-modified Michigan alcoholism screening test (Mm-MAST) and bulimic symptoms [eating disorder inventory-2, bulimia subscale (EDI-Bulimia), population and clinical scoring systems] were defined at age 22-27. We examined relationships between SSS, Mm-MAST, and EDI-Bulimia using Pearson's correlation coefficient (r) and linear regression. RESULTS Alcohol problems were moderately correlated with sensation seeking and bulimic symptoms (population scoring) among women and men (r = 0.21-0.31). The correlation between sensation seeking and bulimic symptoms (population scoring) was weak among men (r = 0.06, p = 0.006) and even weaker and non-significant among women (r = 0.03, p = 0.214). Adjustment for alcohol problems removed the association between sensation seeking and bulimic symptoms among men. Furthermore, there were no significant correlations between sensation seeking and bulimic symptoms when assessing EDI-Bulimia clinical scoring. CONCLUSION Sensation seeking and bulimic symptoms were not associated among women. The association between sensation seeking and bulimic symptoms among men was entirely attributable to increased alcohol problems among those with higher sensation seeking. While this association may be important on the population level, its clinical significance may be minor. LEVEL OF EVIDENCE Level III, well-designed cohort study.
Collapse
Affiliation(s)
- Leon Hirvelä
- Department of Public Health, University of Helsinki, Helsinki, Finland.
| | - Pyry N Sipilä
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | | |
Collapse
|
29
|
Nustad HE, Steinsland I, Ollikainen M, Cazaly E, Kaprio J, Benjamini Y, Gervin K, Lyle R. Modeling dependency structures in 450k DNA methylation data. Bioinformatics 2022; 38:885-891. [PMID: 34788815 PMCID: PMC8796368 DOI: 10.1093/bioinformatics/btab774] [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: 04/19/2021] [Revised: 11/01/2021] [Accepted: 11/09/2021] [Indexed: 02/04/2023] Open
Abstract
MOTIVATION DNA methylation has been shown to be spatially dependent across chromosomes. Previous studies have focused on the influence of genomic context on the dependency structure, while not considering differences in dependency structure between individuals. RESULTS We modeled spatial dependency with a flexible framework to quantify the dependency structure, focusing on inter-individual differences by exploring the association between dependency parameters and technical and biological variables. The model was applied to a subset of the Finnish Twin Cohort study (N = 1611 individuals). The estimates of the dependency parameters varied considerably across individuals, but were generally consistent across chromosomes within individuals. The variation in dependency parameters was associated with bisulfite conversion plate, zygosity, sex and age. The age differences presumably reflect accumulated environmental exposures and/or accumulated small methylation differences caused by stochastic mitotic events, establishing recognizable, individual patterns more strongly seen in older individuals. AVAILABILITY AND IMPLEMENTATION The twin dataset used in the current study are located in the Biobank of the National Institute for Health and Welfare, Finland. All the biobanked data are publicly available for use by qualified researchers following a standardized application procedure (https://thl.fi/en/web/thl-biobank/for-researchers). A R-script for fitting the dependency structure to publicly available DNA methylation data with the software used in this article is provided in supplementary data. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Haakon E Nustad
- Department of Medical Genetics and Norwegian Sequencing Centre, Oslo University Hospital, 0450 Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- Department of Pharmacy, PharmaTox Strategic Research Initiative, University of Oslo, 0371 Oslo, Norway
- Centre for Fertility and Health, Norwegian Institute of Public Health, 0213 Oslo, Norway
| | - Ingelin Steinsland
- Department of Mathematical Sciences, Norwegian University of Science and Technology, 7034 Trondheim, Norway
| | - Miina Ollikainen
- Institute for Molecular Medicine Finland FIMM, Helsinki Institute of Life Science, University of Helsinki, FI-00014 Helsinki, Finland
| | - Emma Cazaly
- Institute for Molecular Medicine Finland FIMM, Helsinki Institute of Life Science, University of Helsinki, FI-00014 Helsinki, Finland
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland FIMM, Helsinki Institute of Life Science, University of Helsinki, FI-00014 Helsinki, Finland
| | - Yuval Benjamini
- Department of Statistics and Data Science, The Hebrew University, Mount Scopus, Jerusalem 9190501, Israel
| | - Kristina Gervin
- Department of Pharmacy, PharmaTox Strategic Research Initiative, University of Oslo, 0371 Oslo, Norway
- Division of Clinical Neuroscience, Department of Research and Innovation, Oslo University Hospital, 0450 Oslo, Norway
- Pharmacoepidemiology and Drug Safety Research Group, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, 0363 Oslo, Norway
| | - Robert Lyle
- Department of Medical Genetics and Norwegian Sequencing Centre, Oslo University Hospital, 0450 Oslo, Norway
- Centre for Fertility and Health, Norwegian Institute of Public Health, 0213 Oslo, Norway
| |
Collapse
|
30
|
Kankaanpää A, Tolvanen A, Saikkonen P, Heikkinen A, Laakkonen EK, Kaprio J, Ollikainen M, Sillanpää E. Do epigenetic clocks provide explanations for sex differences in lifespan? A cross-sectional twin study. J Gerontol A Biol Sci Med Sci 2021; 77:1898-1906. [PMID: 34752604 PMCID: PMC9434475 DOI: 10.1093/gerona/glab337] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Indexed: 11/22/2022] Open
Abstract
Background The sex gap in life expectancy has been narrowing in Finland over the past 4–5 decades; however, on average, women still live longer than men. Epigenetic clocks are markers for biological aging which predict life span. In this study, we examined the mediating role of lifestyle factors on the association between sex and biological aging in younger and older adults. Methods Our sample consists of younger and older twins (21‒42 years, n = 1 477; 50‒76 years, n = 763) including 151 complete younger opposite-sex twin pairs (21‒30 years). Blood-based DNA methylation was used to compute epigenetic age acceleration by 4 epigenetic clocks as a measure of biological aging. Path modeling was used to study whether the association between sex and biological aging is mediated through lifestyle-related factors, that is, education, body mass index, smoking, alcohol use, and physical activity. Results In comparison to women, men were biologically older and, in general, they had unhealthier life habits. The effect of sex on biological aging was partly mediated by body mass index and, in older twins, by smoking. Sex was directly associated with biological aging and the association was stronger in older twins. Conclusions Previously reported sex differences in life span are also evident in biological aging. Declining smoking prevalence among men is a plausible explanation for the narrowing of the difference in life expectancy between the sexes. Data generated by the epigenetic clocks may help in estimating the effects of lifestyle and environmental factors on aging and in predicting aging in future generations.
Collapse
Affiliation(s)
- Anna Kankaanpää
- Gerontology Research Center (GEREC), Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Asko Tolvanen
- Methodology Center for Human Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Pirkko Saikkonen
- Gerontology Research Center (GEREC), Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Aino Heikkinen
- Institute for Molecular Medicine Finland (FIMM), HiLife, University of Helsinki, Helsinki, Finland
| | - Eija K Laakkonen
- Gerontology Research Center (GEREC), Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland (FIMM), HiLife, University of Helsinki, Helsinki, Finland
| | - Miina Ollikainen
- Institute for Molecular Medicine Finland (FIMM), HiLife, University of Helsinki, Helsinki, Finland.,Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Elina Sillanpää
- Gerontology Research Center (GEREC), Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland.,Institute for Molecular Medicine Finland (FIMM), HiLife, University of Helsinki, Helsinki, Finland
| |
Collapse
|
31
|
Lymphoma-Associated Biomarkers Are Increased in Current Smokers in Twin Pairs Discordant for Smoking. Cancers (Basel) 2021; 13:cancers13215395. [PMID: 34771561 PMCID: PMC8582438 DOI: 10.3390/cancers13215395] [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: 09/17/2021] [Accepted: 10/19/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Smoking is associated with a moderate increased risk of Hodgkin and follicular lymphoma. To help understand why, we examined lymphoma-related biomarker levels among 134 smoking and non-smoking twins (67 pairs) ascertained from the Finnish Twin Cohort. We validated self-reported smoking history by measuring serum cotinine, a metabolite of nicotine, from previously collected frozen serum samples. In total, 27 immune biomarkers were assayed using the Luminex Multiplex platform (R & D Systems). We found that four immune response biomarkers were higher and one was lower among smoking compared to non-smoking twins. The strongest association was observed for CCL17/TARC, a biomarker elevated in Hodgkin lymphoma patients. Immune biomarker levels were similar in former smokers and non-smokers. Current smoking may increase levels of immune proteins that could partially explain the association between smoking and risk of certain lymphomas. Abstract Smoking is associated with a moderate increased risk of Hodgkin and follicular lymphoma. To understand why, we examined lymphoma-related biomarker levels among 134 smoking and non-smoking twins (67 pairs) ascertained from the Finnish Twin Cohort. Previously collected frozen serum samples were tested for cotinine to validate self-reported smoking history. In total, 27 immune biomarkers were assayed using the Luminex Multiplex platform (R & D Systems). Current and non-current smokers were defined by a serum cotinine concentration of >3.08 ng/mL and ≤3.08 ng/mL, respectively. Associations between biomarkers and smoking were assessed using linear mixed models to estimate beta coefficients and standard errors, adjusting for age, sex and twin pair as a random effect. There were 55 never smokers, 43 current smokers and 36 former smokers. CCL17/TARC, sgp130, haptoglobin, B-cell activating factor (BAFF) and monocyte chemoattractant protein-1 (MCP1) were significantly (p < 0.05) associated with current smoking and correlated with increasing cotinine concentrations (Ptrend < 0.05). The strongest association was observed for CCL17/TARC (Ptrend = 0.0001). Immune biomarker levels were similar in former and never smokers. Current smoking is associated with increased levels of lymphoma-associated biomarkers, suggesting a possible mechanism for the link between smoking and risk of these two B-cell lymphomas.
Collapse
|
32
|
van Dongen J, Gordon SD, McRae AF, Odintsova VV, Mbarek H, Breeze CE, Sugden K, Lundgren S, Castillo-Fernandez JE, Hannon E, Moffitt TE, Hagenbeek FA, van Beijsterveldt CEM, Jan Hottenga J, Tsai PC, Min JL, Hemani G, Ehli EA, Paul F, Stern CD, Heijmans BT, Slagboom PE, Daxinger L, van der Maarel SM, de Geus EJC, Willemsen G, Montgomery GW, Reversade B, Ollikainen M, Kaprio J, Spector TD, Bell JT, Mill J, Caspi A, Martin NG, Boomsma DI. Identical twins carry a persistent epigenetic signature of early genome programming. Nat Commun 2021; 12:5618. [PMID: 34584077 PMCID: PMC8479069 DOI: 10.1038/s41467-021-25583-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 07/19/2021] [Indexed: 02/08/2023] Open
Abstract
Monozygotic (MZ) twins and higher-order multiples arise when a zygote splits during pre-implantation stages of development. The mechanisms underpinning this event have remained a mystery. Because MZ twinning rarely runs in families, the leading hypothesis is that it occurs at random. Here, we show that MZ twinning is strongly associated with a stable DNA methylation signature in adult somatic tissues. This signature spans regions near telomeres and centromeres, Polycomb-repressed regions and heterochromatin, genes involved in cell-adhesion, WNT signaling, cell fate, and putative human metastable epialleles. Our study also demonstrates a never-anticipated corollary: because identical twins keep a lifelong molecular signature, we can retrospectively diagnose if a person was conceived as monozygotic twin.
Collapse
Affiliation(s)
- Jenny van Dongen
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
- Amsterdam Reproduction and Development (AR&D) Research Institute, Amsterdam, The Netherlands.
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands.
| | - Scott D Gordon
- Queensland Institute of Medical Research Berghofer, Brisbane, QLD, Australia
| | - Allan F McRae
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Veronika V Odintsova
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development (AR&D) Research Institute, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Hamdi Mbarek
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development (AR&D) Research Institute, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | | | - Karen Sugden
- Department of Psychology and Neuroscience and Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Sara Lundgren
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland
| | | | - Eilis Hannon
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Terrie E Moffitt
- Department of Psychology and Neuroscience and Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
- Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Fiona A Hagenbeek
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Catharina E M van Beijsterveldt
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Jouke Jan Hottenga
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Pei-Chien Tsai
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, UK
| | - Josine L Min
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - Gibran Hemani
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - Erik A Ehli
- Avera Institute for Human Genetics, Sioux Falls, SD, USA
| | - Franziska Paul
- Institute of Molecular and Cellular Biology, A*STAR, Singapore, Singapore
| | - Claudio D Stern
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Bastiaan T Heijmans
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - P Eline Slagboom
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Lucia Daxinger
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Eco J C de Geus
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Gonneke Willemsen
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Grant W Montgomery
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Bruno Reversade
- Institute of Molecular and Cellular Biology, A*STAR, Singapore, Singapore
- Genome Institute of Singapore, A*STAR, Singapore, Singapore
- Medical Genetics Department, KOC University, School of Medicine, Istanbul, Turkey
| | - Miina Ollikainen
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, UK
| | - Jordana T Bell
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, UK
| | - Jonathan Mill
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Avshalom Caspi
- Department of Psychology and Neuroscience and Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
- Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Nicholas G Martin
- Queensland Institute of Medical Research Berghofer, Brisbane, QLD, Australia
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development (AR&D) Research Institute, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| |
Collapse
|
33
|
Abtahi H, Gholamzadeh M, Shahmoradi L, Shariat M. An information-based framework for development national twin registry: Scoping review and focus group discussion. Int J Health Plann Manage 2021; 36:1423-1444. [PMID: 34519094 DOI: 10.1002/hpm.3256] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 04/26/2021] [Accepted: 05/18/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Registries in various clinical domains have been established in the last decades. The specific genetic structure of twins has enabled researchers to find answers to the role of genetics and the environment in medical sciences. Thus, twin registries were developed across the world to support twin studies. Our main objective was to devise a conceptual model for developing the national twin registry to ensure the success of this registry. METHODS In this descriptive and qualitative study, the combination of literature review and focus group discussions was applied to achieve suitable models for developing a national twin registry based on lessons learned from founded registries. The qualitative synthesis and reporting results were conducted based on the COREQ checklist. RESULTS According to a systematic literature review, the characteristics and employed strategies employed by established twin registries were recognized. Moreover, based on our objectives, suitable models for registry development were defined. The source of information, the different levels of data, and the information flow were determined based on this model. CONCLUSION Suggesting a conceptual framework for twin registry development at the national level based on the experiences of other countries could contribute to a greater understanding of twin registry implementation efficiently.
Collapse
Affiliation(s)
- Hamidreza Abtahi
- Associate Professor of Pulmonary and Critical Care Department, Thoracic Research Center, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Marsa Gholamzadeh
- Ph.D. Student in Medical Informatics, Health Information Management Department, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Shahmoradi
- Associate Professor of Health Information Management, Health Information Management Department, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Mamak Shariat
- Family Health Research Institute, Maternal-Fetal and Neonatal Research Center, Tehran University of Medical Science, Tehran, Iran
| |
Collapse
|
34
|
Berntzen BJ, Paavonen EJ, Rissanen A, Kaprio J, Pietiläinen KH. Sleep and lifestyle in young adult monozygotic twin pairs discordant for body mass index. Sleep Health 2021; 7:556-564. [PMID: 34193396 DOI: 10.1016/j.sleh.2021.04.002] [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: 10/05/2020] [Revised: 04/07/2021] [Accepted: 04/13/2021] [Indexed: 11/19/2022]
Abstract
OBJECTIVES The causal nature of the sleep-obesity association is unclear. To control for potential confounding by genes and shared environment, we studied monozygotic twin pairs discordant for body mass index (BMI). First, we investigated sleep in relation to BMI. Second, we examined associations of objective and subjective sleep duration and sleep debt (objective or subjective sleep duration minus subjective sleep need) with eating behaviors and physical activity (PA). DESIGN Cross-sectional study. SETTING Finnish twins in everyday life circumstances. PARTICIPANTS Seventy-four healthy young adult monozygotic twin pairs, of whom 36 were BMI-discordant (∆BMI ≥ 3 kg/m2). MEASUREMENTS Clinical measurements estimated BMI and body composition. Sleep, eating, and PA behaviors were measured by self-report and actigraphy. RESULTS Compared to co-twins with lower BMI, co-twins with higher BMI reported shorter sleep (P = .043), more snoring (P = .0093), and greater tiredness (P = .0013) and trended toward eveningness (P = .036). Actigraphy-measured sleep duration correlated highly within BMI-discordant twin pairs (r = 0.63, P = .004). Subjective sleep debt was consistently positively associated with disinhibited eating and binge eating, but not with BMI. Subjective and objective sleep debt had negative correlations with moderate-to-vigorous PA. CONCLUSIONS Twins with higher BMI showed less favorable sleep characteristics than their co-twins with lower BMI. Subjective sleep debt is a potential target for intervention to reduce eating and PA behaviors that promote weight gain. Experimental studies could elucidate mechanisms underlying tiredness in individuals with higher BMI and investigate causal relationships between sleep debt, BMI, and lifestyle.
Collapse
Affiliation(s)
- Bram J Berntzen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - E Juulia Paavonen
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland; Pediatric Research Center, Child Psychiatry, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Aila Rissanen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland; Department of Public Health, Finnish Twin Cohort Study, University of Helsinki, Helsinki, Finland
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Obesity Center, Endocrinology, Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
35
|
Sillanpää E, Heikkinen A, Kankaanpää A, Paavilainen A, Kujala UM, Tammelin TH, Kovanen V, Sipilä S, Pietiläinen KH, Kaprio J, Ollikainen M, Laakkonen EK. Blood and skeletal muscle ageing determined by epigenetic clocks and their associations with physical activity and functioning. Clin Epigenetics 2021; 13:110. [PMID: 34001218 PMCID: PMC8127311 DOI: 10.1186/s13148-021-01094-6] [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: 02/12/2021] [Accepted: 04/26/2021] [Indexed: 12/14/2022] Open
Abstract
The aim of this study was to investigate the correspondence of different biological ageing estimates (i.e. epigenetic age) in blood and muscle tissue and their associations with physical activity (PA), physical function and body composition. Two independent cohorts (N = 139 and N = 47) were included, whose age span covered adulthood (23–69 years). Whole blood and m. vastus lateralis samples were collected, and DNA methylation was analysed. Four different DNA methylation age (DNAmAge) estimates were calculated using genome-wide methylation data and publicly available online tools. A novel muscle-specific methylation age was estimated using the R-package ‘MEAT’. PA was measured with questionnaires and accelerometers. Several tests were conducted to estimate cardiorespiratory fitness and muscle strength. Body composition was estimated by dual-energy X-ray absorptiometry. DNAmAge estimates from blood and muscle were highly correlated with chronological age, but different age acceleration estimates were weakly associated with each other. The monozygotic twin within-pair similarity of ageing pace was higher in blood (r = 0.617–0.824) than in muscle (r = 0.523–0.585). Associations of age acceleration estimates with PA, physical function and body composition were weak in both tissues and mostly explained by smoking and sex. The muscle-specific epigenetic clock MEAT was developed to predict chronological age, which may explain why it did not associate with functional phenotypes. The Horvath’s clock and GrimAge were weakly associated with PA and related phenotypes, suggesting that higher PA would be linked to accelerated biological ageing in muscle. This may, however, be more reflective of the low capacity of epigenetic clock algorithms to measure functional muscle ageing than of actual age acceleration. Based on our results, the investigated epigenetic clocks have rather low value in estimating muscle ageing with respect to the physiological adaptations that typically occur due to ageing or PA. Thus, further development of methods is needed to gain insight into muscle tissue-specific ageing and the underlying biological pathways.
Collapse
Affiliation(s)
- Elina Sillanpää
- Gerontology Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, P.O. Box 35 (VIV), 40014, Jyväskylä, Finland. .,Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland.
| | - Aino Heikkinen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland.,Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Anna Kankaanpää
- Gerontology Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, P.O. Box 35 (VIV), 40014, Jyväskylä, Finland
| | - Aini Paavilainen
- Gerontology Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, P.O. Box 35 (VIV), 40014, Jyväskylä, Finland
| | - Urho M Kujala
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Tuija H Tammelin
- LIKES Research Centre for Physical Activity and Health, Jyväskylä, Finland
| | - Vuokko Kovanen
- Gerontology Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, P.O. Box 35 (VIV), 40014, Jyväskylä, Finland
| | - Sarianna Sipilä
- Gerontology Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, P.O. Box 35 (VIV), 40014, Jyväskylä, Finland
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Obesity Center, Endocrinology, Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Miina Ollikainen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland.,Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Eija K Laakkonen
- Gerontology Research Center, Faculty of Sport and Health Sciences, University of Jyväskylä, P.O. Box 35 (VIV), 40014, Jyväskylä, Finland
| |
Collapse
|
36
|
Molecular pathways behind acquired obesity: Adipose tissue and skeletal muscle multiomics in monozygotic twin pairs discordant for BMI. CELL REPORTS MEDICINE 2021; 2:100226. [PMID: 33948567 PMCID: PMC8080113 DOI: 10.1016/j.xcrm.2021.100226] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/31/2020] [Accepted: 03/04/2021] [Indexed: 12/12/2022]
Abstract
Tissue-specific mechanisms prompting obesity-related development complications in humans remain unclear. We apply multiomics analyses of subcutaneous adipose tissue and skeletal muscle to examine the effects of acquired obesity among 49 BMI-discordant monozygotic twin pairs. Overall, adipose tissue appears to be more affected by excess body weight than skeletal muscle. In heavier co-twins, we observe a transcriptional pattern of downregulated mitochondrial pathways in both tissues and upregulated inflammatory pathways in adipose tissue. In adipose tissue, heavier co-twins exhibit lower creatine levels; in skeletal muscle, glycolysis- and redox stress-related protein and metabolite levels remain higher. Furthermore, metabolomics analyses in both tissues reveal that several proinflammatory lipids are higher and six of the same lipid derivatives are lower in acquired obesity. Finally, in adipose tissue, but not in skeletal muscle, mitochondrial downregulation and upregulated inflammation are associated with a fatty liver, insulin resistance, and dyslipidemia, suggesting that adipose tissue dominates in acquired obesity. Multiomics analyses of adipose tissue and skeletal muscle in BMI-discordant twins Excess body weight downregulates mitochondrial pathways in both tissues Excess body weight upregulates proinflammatory pathways in both tissues Adipose tissue alterations are associated with metabolic health in acquired obesity
Collapse
|
37
|
Heiskanen MA, Honkala SM, Hentilä J, Ojala R, Lautamäki R, Koskensalo K, Lietzén MS, Saunavaara V, Saunavaara J, Helmiö M, Löyttyniemi E, Nummenmaa L, Collado MC, Malm T, Lahti L, Pietiläinen KH, Kaprio J, Rinne JO, Hannukainen JC. Systemic cross-talk between brain, gut, and peripheral tissues in glucose homeostasis: effects of exercise training (CROSSYS). Exercise training intervention in monozygotic twins discordant for body weight. BMC Sports Sci Med Rehabil 2021; 13:16. [PMID: 33627179 PMCID: PMC7905681 DOI: 10.1186/s13102-021-00241-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 02/03/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Obesity and physical inactivity are major global public health concerns, both of which increase the risk of insulin resistance and type 2 diabetes. Regulation of glucose homeostasis involves cross-talk between the central nervous system, peripheral tissues, and gut microbiota, and is affected by genetics. Systemic cross-talk between brain, gut, and peripheral tissues in glucose homeostasis: effects of exercise training (CROSSYS) aims to gain new systems-level understanding of the central metabolism in human body, and how exercise training affects this cross-talk. METHODS CROSSYS is an exercise training intervention, in which participants are monozygotic twins from pairs discordant for body mass index (BMI) and within a pair at least the other is overweight. Twins are recruited from three population-based longitudinal Finnish twin studies, including twins born in 1983-1987, 1975-1979, and 1945-1958. The participants undergo 6-month-long exercise intervention period, exercising four times a week (including endurance, strength, and high-intensity training). Before and after the exercise intervention, comprehensive measurements are performed in Turku PET Centre, Turku, Finland. The measurements include: two positron emission tomography studies (insulin-stimulated whole-body and tissue-specific glucose uptake and neuroinflammation), magnetic resonance imaging (brain morphology and function, quantification of body fat masses and organ volumes), magnetic resonance spectroscopy (quantification of fat within heart, pancreas, liver and tibialis anterior muscle), echocardiography, skeletal muscle and adipose tissue biopsies, a neuropsychological test battery as well as biosamples from blood, urine and stool. The participants also perform a maximal exercise capacity test and tests of muscular strength. DISCUSSION This study addresses the major public health problems related to modern lifestyle, obesity, and physical inactivity. An eminent strength of this project is the possibility to study monozygotic twin pairs that share the genome at the sequence level but are discordant for BMI that is a risk factor for metabolic impairments such as insulin resistance. Thus, this exercise training intervention elucidates the effects of obesity on metabolism and whether regular exercise training is able to reverse obesity-related impairments in metabolism in the absence of the confounding effects of genetic factors. TRIAL REGISTRATION ClinicalTrials.gov , NCT03730610 . Prospectively registered 5 November 2018.
Collapse
Affiliation(s)
- Marja A Heiskanen
- Turku PET Centre, University of Turku, P.O. Box 52, FIN-20521, Turku, Finland
| | - Sanna M Honkala
- Turku PET Centre, University of Turku, P.O. Box 52, FIN-20521, Turku, Finland
| | - Jaakko Hentilä
- Turku PET Centre, University of Turku, P.O. Box 52, FIN-20521, Turku, Finland
| | - Ronja Ojala
- Turku PET Centre, University of Turku, P.O. Box 52, FIN-20521, Turku, Finland
| | | | - Kalle Koskensalo
- Department of Medical Physics, Turku University Hospital, Turku, Finland
| | - Martin S Lietzén
- Turku PET Centre, University of Turku, P.O. Box 52, FIN-20521, Turku, Finland
| | - Virva Saunavaara
- Turku PET Centre, University of Turku, P.O. Box 52, FIN-20521, Turku, Finland
- Department of Medical Physics, Turku University Hospital, Turku, Finland
| | - Jani Saunavaara
- Department of Medical Physics, Turku University Hospital, Turku, Finland
| | - Mika Helmiö
- Division of Digestive Surgery and Urology, Turku University Hospital, Turku, Finland
| | | | - Lauri Nummenmaa
- Turku PET Centre, University of Turku, P.O. Box 52, FIN-20521, Turku, Finland
- Department of Psychology, University of Turku, Turku, Finland
| | - Maria C Collado
- Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Valencia, Spain
- Functional Food Forum, University of Turku, Turku, Finland
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Leo Lahti
- Department of Future Technologies, University of Turku, Turku, Finland
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Abdominal Center, Obesity Center, Endocrinology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland FIMM, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Juha O Rinne
- Turku PET Centre, University of Turku, P.O. Box 52, FIN-20521, Turku, Finland
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Jarna C Hannukainen
- Turku PET Centre, University of Turku, P.O. Box 52, FIN-20521, Turku, Finland.
| |
Collapse
|
38
|
Masip G, Silventoinen K, Keski-Rahkonen A, Palviainen T, Sipilä PN, Kaprio J, Bogl LH. The genetic architecture of the association between eating behaviors and obesity: combining genetic twin modeling and polygenic risk scores. Am J Clin Nutr 2020; 112:956-966. [PMID: 32685959 PMCID: PMC7528566 DOI: 10.1093/ajcn/nqaa181] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 06/12/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Obesity susceptibility genes are highly expressed in the brain suggesting that they might exert their influence on body weight through eating-related behaviors. OBJECTIVES To examine whether the genetic susceptibility to obesity is mediated by eating behavior patterns. METHODS Participants were 3977 twins (33% monozygotic, 56% females), aged 31-37 y, from wave 5 of the FinnTwin16 study. They self-reported their height and weight, eating behaviors (15 items), diet quality, and self-measured their waist circumference (WC). For 1055 twins with genome-wide data, we constructed a polygenic risk score for BMI (PRSBMI) using almost 1 million single nucleotide polymorphisms. We used principal component analyses to identify eating behavior patterns, twin modeling to decompose correlations into genetic and environmental components, and structural equation modeling to test mediation models between the PRSBMI, eating behavior patterns, and obesity measures. RESULTS We identified 4 moderately heritable (h2 = 36-48%) eating behavior patterns labeled "snacking," "infrequent and unhealthy eating," "avoidant eating," and "emotional and external eating." The highest phenotypic correlation with obesity measures was found for the snacking behavior pattern (r = 0.35 for BMI and r = 0.32 for WC; P < 0.001 for both), largely due to genetic factors in common (bivariate h2 > 70%). The snacking behavior pattern partially mediated the association between the PRSBMI and obesity measures (βindirect = 0.06; 95% CI: 0.02, 0.09; P = 0.002 for BMI; and βindirect = 0.05; 95% CI: 0.02, 0.08; P = 0.003 for WC). CONCLUSIONS Eating behavior patterns share a common genetic liability with obesity measures and are moderately heritable. Genetic susceptibility to obesity can be partly mediated by an eating pattern characterized by frequent snacking. Obesity prevention efforts might therefore benefit from focusing on eating behavior change, particularly in genetically susceptible individuals.
Collapse
Affiliation(s)
- Guiomar Masip
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Karri Silventoinen
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Population Research Unit, Faculty of Social Sciences, University of Helsinki, Helsinki, Finland
| | | | - Teemu Palviainen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Pyry N Sipilä
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Jaakko Kaprio
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Leonie H Bogl
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Department of Epidemiology, Center for Public Health, Medical University of Vienna, Vienna, Austria
| |
Collapse
|