Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Andrea Ganna is active.

Publication


Featured researches published by Andrea Ganna.


Twin Research and Human Genetics | 2013

The Swedish Twin Registry: Establishment of a Biobank and Other Recent Developments

Patrik K. E. Magnusson; Catarina Almqvist; Iffat Rahman; Andrea Ganna; Alexander Viktorin; Hasse Walum; Linda Halldner; Sebastian Lundström; Fredrik Ullén; Niklas Långström; Henrik Larsson; Anastasia Nyman; Clara Hellner Gumpert; Maria Råstam; Henrik Anckarsäter; Sven Cnattingius; Magnus Johannesson; Erik Ingelsson; Lars Klareskog; Ulf de Faire; Nancy L. Pedersen; Paul Lichtenstein

The Swedish Twin Registry (STR) today contains more than 194,000 twins and more than 75,000 pairs have zygosity determined by an intra-pair similarity algorithm, DNA, or by being of opposite sex. Of these, approximately 20,000, 25,000, and 30,000 pairs are monozygotic, same-sex dizygotic, and opposite-sex dizygotic pairs, respectively. Since its establishment in the late 1950s, the STR has been an important epidemiological resource for the study of genetic and environmental influences on a multitude of traits, behaviors, and diseases. Following large investments in the collection of biological specimens in the past 10 years we have now established a Swedish twin biobank with DNA from 45,000 twins and blood serum from 15,000 twins, which effectively has also transformed the registry into a powerful resource for molecular studies. We here describe the main projects within which the new collections of both biological samples as well as phenotypic measures have been collected. Coverage by year of birth, zygosity determination, ethnic heterogeneity, and influences of in vitro fertilization are also described.


The Lancet | 2015

5 year mortality predictors in 498 103 UK Biobank participants: a prospective population-based study

Andrea Ganna; Erik Ingelsson

BACKGROUND To our knowledge, a systematic comparison of predictors of mortality in middle-aged to elderly individuals has not yet been done. We investigated predictors of mortality in UK Biobank participants during a 5 year period. We aimed to investigate the associations between most of the available measurements and 5 year all-cause and cause-specific mortality, and to develop and validate a prediction score for 5 year mortality using only self-reported information. METHODS Participants were enrolled in the UK Biobank from April, 2007, to July, 2010, from 21 assessment centres across England, Wales, and Scotland with standardised procedures. In this prospective population-based study, we assessed sex-specific associations of 655 measurements of demographics, health, and lifestyle with all-cause mortality and six cause-specific mortality categories in UK Biobank participants using the Cox proportional hazard model. We excluded variables that were missing in more than 80% of the participants and all cardiorespiratory fitness test measurements because summary data were not available. Validation of the prediction score was done in participants enrolled at the Scottish centres. UK life tables and census information were used to calibrate the score to the overall UK population. FINDINGS About 500,000 participants were included in the UK Biobank. We excluded participants with more than 80% variables missing (n=746). Of 498,103 UK Biobank participants included (54% of whom were women) aged 37-73 years, 8532 (39% of whom were women) died during a median follow-up of 4·9 years (IQR 4·33-5·22). Self-reported health (C-index including age 0·74 [95% CI 0·73-0·75]) was the strongest predictor of all-cause mortality in men and a previous cancer diagnosis (0·73 [0·72-0·74]) was the strongest predictor of all-cause mortality in women. When excluding individuals with major diseases or disorders (Charlson comorbidity index >0; n=355 043), measures of smoking habits were the strongest predictors of all-cause mortality. The prognostic score including 13 self-reported predictors for men and 11 for women achieved good discrimination (0·80 [0·77-0·83] for men and 0·79 [0·76-0·83] for women) and significantly outperformed the Charlson comorbidity index (p<0·0001 in men and p=0·0007 in women). A dedicated website allows the interactive exploration of all results along with calculation of individual risk through an online questionnaire. INTERPRETATION Measures that can simply be obtained by questionnaires and without physical examination were the strongest predictors of all-cause mortality in the UK Biobank population. The prediction score we have developed accurately predicts 5 year all-cause mortality and can be used by individuals to improve health awareness, and by health professionals and organisations to identify high-risk individuals and guide public policy. FUNDING Knut and Alice Wallenberg Foundation and the Swedish Research Council.


PLOS Genetics | 2013

Gene × Physical Activity Interactions in Obesity: Combined Analysis of 111,421 Individuals of European Ancestry

Shafqat Ahmad; Gull Rukh; Tibor V. Varga; Ashfaq Ali; Azra Kurbasic; Dmitry Shungin; Ulrika Ericson; Robert W. Koivula; Audrey Y. Chu; Lynda M. Rose; Andrea Ganna; Qibin Qi; Alena Stančáková; Camilla H. Sandholt; Cathy E. Elks; Gary C. Curhan; Majken K. Jensen; Rulla M. Tamimi; Kristine H. Allin; Torben Jørgensen; Soren Brage; Claudia Langenberg; Mette Aadahl; Niels Grarup; Allan Linneberg; Guillaume Paré; Patrik K. E. Magnusson; Nancy L. Pedersen; Michael Boehnke; Anders Hamsten

Numerous obesity loci have been identified using genome-wide association studies. A UK study indicated that physical activity may attenuate the cumulative effect of 12 of these loci, but replication studies are lacking. Therefore, we tested whether the aggregate effect of these loci is diminished in adults of European ancestry reporting high levels of physical activity. Twelve obesity-susceptibility loci were genotyped or imputed in 111,421 participants. A genetic risk score (GRS) was calculated by summing the BMI-associated alleles of each genetic variant. Physical activity was assessed using self-administered questionnaires. Multiplicative interactions between the GRS and physical activity on BMI were tested in linear and logistic regression models in each cohort, with adjustment for age, age2, sex, study center (for multicenter studies), and the marginal terms for physical activity and the GRS. These results were combined using meta-analysis weighted by cohort sample size. The meta-analysis yielded a statistically significant GRS × physical activity interaction effect estimate (Pinteraction = 0.015). However, a statistically significant interaction effect was only apparent in North American cohorts (n = 39,810, Pinteraction = 0.014 vs. n = 71,611, Pinteraction = 0.275 for Europeans). In secondary analyses, both the FTO rs1121980 (Pinteraction = 0.003) and the SEC16B rs10913469 (Pinteraction = 0.025) variants showed evidence of SNP × physical activity interactions. This meta-analysis of 111,421 individuals provides further support for an interaction between physical activity and a GRS in obesity disposition, although these findings hinge on the inclusion of cohorts from North America, indicating that these results are either population-specific or non-causal.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2013

Multilocus Genetic Risk Scores for Coronary Heart Disease Prediction

Andrea Ganna; Patrik K. E. Magnusson; Nancy L. Pedersen; Ulf de Faire; Marie Reilly; Johan Ärnlöv; Johan Sundström; Anders Hamsten; Erik Ingelsson

Objective—Current guidelines do not support the use of genetic profiles in risk assessment of coronary heart disease (CHD). However, new single nucleotide polymorphisms associated with CHD and intermediate cardiovascular traits have recently been discovered. We aimed to compare several multilocus genetic risk score (MGRS) in terms of association with CHD and to evaluate clinical use. Approach and Results—We investigated 6 Swedish prospective cohort studies with 10 612 participants free of CHD at baseline. We developed 1 overall MGRS based on 395 single nucleotide polymorphisms reported as being associated with cardiovascular traits, 1 CHD-specific MGRS, including 46 single nucleotide polymorphisms, and 6 trait-specific MGRS for each established CHD risk factors. Both the overall and the CHD-specific MGRS were significantly associated with CHD risk (781 incident events; hazard ratios for fourth versus first quartile, 1.54 and 1.52; P<0.001) and improved risk classification beyond established risk factors (net reclassification improvement, 4.2% and 4.9%; P=0.006 and 0.017). Discrimination improvement was modest (C-index improvement, 0.004). A polygene MGRS performed worse than the CHD-specific MGRS. We estimate that 1 additional CHD event for every 318 people screened at intermediate risk could be saved by measuring the CHD-specific genetic score in addition to the established risk factors. Conclusions—Our results indicate that genetic information could be of some clinical value for prediction of CHD, although further studies are needed to address aspects, such as feasibility, ethics, and cost efficiency of genetic profiling in the primary prevention setting.


PLOS Genetics | 2014

Large-scale metabolomic profiling identifies novel biomarkers for incident coronary heart disease.

Andrea Ganna; Samira Salihovic; Johan Sundström; Corey D. Broeckling; Åsa K. Hedman; Patrik K. E. Magnusson; Nancy L. Pedersen; Anders Larsson; Agneta Siegbahn; Mihkel Zilmer; Jessica E. Prenni; Johan Ärnlöv; Lars Lind; Tove Fall; Erik Ingelsson

Analyses of circulating metabolites in large prospective epidemiological studies could lead to improved prediction and better biological understanding of coronary heart disease (CHD). We performed a mass spectrometry-based non-targeted metabolomics study for association with incident CHD events in 1,028 individuals (131 events; 10 y. median follow-up) with validation in 1,670 individuals (282 events; 3.9 y. median follow-up). Four metabolites were replicated and independent of main cardiovascular risk factors [lysophosphatidylcholine 18∶1 (hazard ratio [HR] per standard deviation [SD] increment = 0.77, P-value<0.001), lysophosphatidylcholine 18∶2 (HR = 0.81, P-value<0.001), monoglyceride 18∶2 (MG 18∶2; HR = 1.18, P-value = 0.011) and sphingomyelin 28∶1 (HR = 0.85, P-value = 0.015)]. Together they contributed to moderate improvements in discrimination and re-classification in addition to traditional risk factors (C-statistic: 0.76 vs. 0.75; NRI: 9.2%). MG 18∶2 was associated with CHD independently of triglycerides. Lysophosphatidylcholines were negatively associated with body mass index, C-reactive protein and with less evidence of subclinical cardiovascular disease in additional 970 participants; a reverse pattern was observed for MG 18∶2. MG 18∶2 showed an enrichment (P-value = 0.002) of significant associations with CHD-associated SNPs (P-value = 1.2×10−7 for association with rs964184 in the ZNF259/APOA5 region) and a weak, but positive causal effect (odds ratio = 1.05 per SD increment in MG 18∶2, P-value = 0.05) on CHD, as suggested by Mendelian randomization analysis. In conclusion, we identified four lipid-related metabolites with evidence for clinical utility, as well as a causal role in CHD development.


American Journal of Epidemiology | 2013

Meta-Analysis Investigating Associations Between Healthy Diet and Fasting Glucose and Insulin Levels and Modification by Loci Associated With Glucose Homeostasis in Data From 15 Cohorts

Jennifer A. Nettleton; Marie-France Hivert; Rozenn N. Lemaitre; Nicola M. McKeown; Dariush Mozaffarian; Toshiko Tanaka; Mary K. Wojczynski; Adela Hruby; Luc Djoussé; Julius S. Ngwa; Jack L. Follis; Maria Dimitriou; Andrea Ganna; Denise K. Houston; Stavroula Kanoni; Vera Mikkilä; Ani Manichaikul; Ioanna Ntalla; Frida Renström; Emily Sonestedt; Frank J. A. van Rooij; Stefania Bandinelli; Lawrence de Koning; Ulrika Ericson; Neelam Hassanali; Jessica C. Kiefte-de Jong; Kurt Lohman; Olli T. Raitakari; Constantina Papoutsakis; Per Sjögren

Whether loci that influence fasting glucose (FG) and fasting insulin (FI) levels, as identified by genome-wide association studies, modify associations of diet with FG or FI is unknown. We utilized data from 15 U.S. and European cohort studies comprising 51,289 persons without diabetes to test whether genotype and diet interact to influence FG or FI concentration. We constructed a diet score using study-specific quartile rankings for intakes of whole grains, fish, fruits, vegetables, and nuts/seeds (favorable) and red/processed meats, sweets, sugared beverages, and fried potatoes (unfavorable). We used linear regression within studies, followed by inverse-variance-weighted meta-analysis, to quantify 1) associations of diet score with FG and FI levels and 2) interactions of diet score with 16 FG-associated loci and 2 FI-associated loci. Diet score (per unit increase) was inversely associated with FG (β = -0.004 mmol/L, 95% confidence interval: -0.005, -0.003) and FI (β = -0.008 ln-pmol/L, 95% confidence interval: -0.009, -0.007) levels after adjustment for demographic factors, lifestyle, and body mass index. Genotype variation at the studied loci did not modify these associations. Healthier diets were associated with lower FG and FI concentrations regardless of genotype at previously replicated FG- and FI-associated loci. Studies focusing on genomic regions that do not yield highly statistically significant associations from main-effect genome-wide association studies may be more fruitful in identifying diet-gene interactions.


Journal of Nutrition | 2013

Higher Magnesium Intake Is Associated with Lower Fasting Glucose and Insulin, with No Evidence of Interaction with Select Genetic Loci, in a Meta-Analysis of 15 CHARGE Consortium Studies

Adela Hruby; Julius S. Ngwa; Frida Renström; Mary K. Wojczynski; Andrea Ganna; Göran Hallmans; Denise K. Houston; Paul F. Jacques; Stavroula Kanoni; Terho Lehtimäki; Rozenn N. Lemaitre; Ani Manichaikul; Kari E. North; Ioanna Ntalla; Emily Sonestedt; Toshiko Tanaka; Frank J. A. van Rooij; Stefania Bandinelli; Luc Djoussé; Efi Grigoriou; Ingegerd Johansson; Kurt Lohman; James S. Pankow; Olli T. Raitakari; Ulf Risérus; Mary Yannakoulia; M. Carola Zillikens; Neelam Hassanali; Yongmei Liu; Dariush Mozaffarian

Favorable associations between magnesium intake and glycemic traits, such as fasting glucose and insulin, are observed in observational and clinical studies, but whether genetic variation affects these associations is largely unknown. We hypothesized that single nucleotide polymorphisms (SNPs) associated with either glycemic traits or magnesium metabolism affect the association between magnesium intake and fasting glucose and insulin. Fifteen studies from the CHARGE (Cohorts for Heart and Aging Research in Genomic Epidemiology) Consortium provided data from up to 52,684 participants of European descent without known diabetes. In fixed-effects meta-analyses, we quantified 1) cross-sectional associations of dietary magnesium intake with fasting glucose (mmol/L) and insulin (ln-pmol/L) and 2) interactions between magnesium intake and SNPs related to fasting glucose (16 SNPs), insulin (2 SNPs), or magnesium (8 SNPs) on fasting glucose and insulin. After adjustment for age, sex, energy intake, BMI, and behavioral risk factors, magnesium (per 50-mg/d increment) was inversely associated with fasting glucose [β = -0.009 mmol/L (95% CI: -0.013, -0.005), P < 0.0001] and insulin [-0.020 ln-pmol/L (95% CI: -0.024, -0.017), P < 0.0001]. No magnesium-related SNP or interaction between any SNP and magnesium reached significance after correction for multiple testing. However, rs2274924 in magnesium transporter-encoding TRPM6 showed a nominal association (uncorrected P = 0.03) with glucose, and rs11558471 in SLC30A8 and rs3740393 near CNNM2 showed a nominal interaction (uncorrected, both P = 0.02) with magnesium on glucose. Consistent with other studies, a higher magnesium intake was associated with lower fasting glucose and insulin. Nominal evidence of TRPM6 influence and magnesium interaction with select loci suggests that further investigation is warranted.


Human Molecular Genetics | 2015

Gene × dietary pattern interactions in obesity: analysis of up to 68 317 adults of European ancestry

Jennifer A. Nettleton; Jack L. Follis; Julius S. Ngwa; Caren E. Smith; Shafqat Ahmad; Toshiko Tanaka; Mary K. Wojczynski; Trudy Voortman; Rozenn N. Lemaitre; Kati Kristiansson; Marja-Liisa Nuotio; Denise K. Houston; Mia-Maria Perälä; Qibin Qi; Emily Sonestedt; Ani Manichaikul; Stavroula Kanoni; Andrea Ganna; Vera Mikkilä; Kari E. North; David S. Siscovick; Kennet Harald; Nicola M. McKeown; Ingegerd Johansson; Harri Rissanen; Yongmei Liu; Jari Lahti; Frank B. Hu; Stefania Bandinelli; Gull Rukh

Abstract Obesity is highly heritable. Genetic variants showing robust associations with obesity traits have been identified through genome-wide association studies. We investigated whether a composite score representing healthy diet modifies associations of these variants with obesity traits. Totally, 32 body mass index (BMI)- and 14 waist–hip ratio (WHR)-associated single nucleotide polymorphisms were genotyped, and genetic risk scores (GRS) were calculated in 18 cohorts of European ancestry (n = 68 317). Diet score was calculated based on self-reported intakes of whole grains, fish, fruits, vegetables, nuts/seeds (favorable) and red/processed meats, sweets, sugar-sweetened beverages and fried potatoes (unfavorable). Multivariable adjusted, linear regression within each cohort followed by inverse variance-weighted, fixed-effects meta-analysis was used to characterize: (a) associations of each GRS with BMI and BMI-adjusted WHR and (b) diet score modification of genetic associations with BMI and BMI-adjusted WHR. Nominally significant interactions (P = 0.006–0.04) were observed between the diet score and WHR-GRS (but not BMI-GRS), two WHR loci (GRB14 rs10195252; LYPLAL1 rs4846567) and two BMI loci (LRRN6C rs10968576; MTIF3 rs4771122), for the respective BMI-adjusted WHR or BMI outcomes. Although the magnitudes of these select interactions were small, our data indicated that associations between genetic predisposition and obesity traits were stronger with a healthier diet. Our findings generate interesting hypotheses; however, experimental and functional studies are needed to determine their clinical relevance.


Nature | 2017

The impact of rare variation on gene expression across tissues

Xin Li; Yungil Kim; Emily K. Tsang; Joe R. Davis; Farhan N. Damani; Colby Chiang; Gaelen T. Hess; Zachary Zappala; Benjamin J. Strober; Alexandra J. Scott; Amy Li; Andrea Ganna; Michael C. Bassik; Jason D. Merker; Ira M. Hall; Alexis Battle; Stephen B. Montgomery

Rare genetic variants are abundant in humans and are expected to contribute to individual disease risk. While genetic association studies have successfully identified common genetic variants associated with susceptibility, these studies are not practical for identifying rare variants. Efforts to distinguish pathogenic variants from benign rare variants have leveraged the genetic code to identify deleterious protein-coding alleles, but no analogous code exists for non-coding variants. Therefore, ascertaining which rare variants have phenotypic effects remains a major challenge. Rare non-coding variants have been associated with extreme gene expression in studies using single tissues, but their effects across tissues are unknown. Here we identify gene expression outliers, or individuals showing extreme expression levels for a particular gene, across 44 human tissues by using combined analyses of whole genomes and multi-tissue RNA-sequencing data from the Genotype-Tissue Expression (GTEx) project v6p release. We find that 58% of underexpression and 28% of overexpression outliers have nearby conserved rare variants compared to 8% of non-outliers. Additionally, we developed RIVER (RNA-informed variant effect on regulation), a Bayesian statistical model that incorporates expression data to predict a regulatory effect for rare variants with higher accuracy than models using genomic annotations alone. Overall, we demonstrate that rare variants contribute to large gene expression changes across tissues and provide an integrative method for interpretation of rare variants in individual genomes.


Nature Neuroscience | 2016

Ultra-rare disruptive and damaging mutations influence educational attainment in the general population

Andrea Ganna; Giulio Genovese; Daniel P. Howrigan; Andrea Byrnes; Mitja I. Kurki; Seyedeh M. Zekavat; Christopher W. Whelan; Mart Kals; Michel G. Nivard; Alex Bloemendal; Jonathan Bloom; Jacqueline I. Goldstein; Timothy Poterba; Cotton Seed; Robert E. Handsaker; Pradeep Natarajan; Reedik Mägi; Diane Gage; Elise B. Robinson; Andres Metspalu; Veikko Salomaa; Jaana Suvisaari; Shaun Purcell; Pamela Sklar; Sekar Kathiresan; Mark J. Daly; Steven A. McCarroll; Patrick F. Sullivan; Aarno Palotie; Tonu Esko

Disruptive, damaging ultra-rare variants in highly constrained genes are enriched in individuals with neurodevelopmental disorders. In the general population, this class of variants was associated with a decrease in years of education (YOE). This effect was stronger among highly brain-expressed genes and explained more YOE variance than pathogenic copy number variation but less than common variants. Disruptive, damaging ultra-rare variants in highly constrained genes influence the determinants of YOE in the general population.

Collaboration


Dive into the Andrea Ganna's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kari E. North

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Veikko Salomaa

National Institute for Health and Welfare

View shared research outputs
Researchain Logo
Decentralizing Knowledge