1
|
Westerman KE, Kilpeläinen TO, Sevilla-Gonzalez M, Connelly MA, Wood AC, Tsai MY, Taylor KD, Rich SS, Rotter JI, Otvos JD, Bentley AR, Mora S, Aschard H, Rao DC, Gu C, Chasman DI, Manning AK. Refinement of a Published Gene-Physical Activity Interaction Impacting HDL-Cholesterol: Role of Sex and Lipoprotein Subfractions. Genet Epidemiol 2025; 49:e22607. [PMID: 39764704 PMCID: PMC11934221 DOI: 10.1002/gepi.22607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 10/09/2024] [Accepted: 12/17/2024] [Indexed: 01/15/2025]
Abstract
Large-scale gene-environment interaction (GxE) discovery efforts often involve analytical compromises for the sake of data harmonization and statistical power. Refinement of exposures, covariates, outcomes, and population subsets may be helpful to establish often-elusive replication and evaluate potential clinical utility. Here, we used additional datasets, an expanded set of statistical models, and interrogation of lipoprotein metabolism via nuclear magnetic resonance (NMR)-based lipoprotein subfractions to refine a previously discovered GxE modifying the relationship between physical activity (PA) and HDL-cholesterol (HDL-C). We explored this GxE in the Women's Genome Health Study (WGHS; N = 23,294; the strongest cohort-specific signal in the original meta-analysis), the UK Biobank (UKB; N = 281,380), and the Multi-Ethnic Study of Atherosclerosis (MESA; N = 4587), using self-reported PA (MET-min/wk) and genotypes at rs295849 (nearest gene: LHX1). As originally reported, minor allele carriers of rs295849 in WGHS had a stronger positive association between PA and HDL-C (pint = 0.002). When testing available NMR metabolites to refine the HDL-C outcome, we found a stronger interaction effect on medium-sized HDL particle concentrations (M-HDL-P; pint = 1.0 × 10-4) than HDL-C. Meta-regression revealed a systematically larger interaction effect in cohorts from the original meta-analysis with a greater fraction of women (p = 0.018). In the UKB, GxE effects were stronger in women and using M-HDL-P as the outcome. In MESA, the primary interaction for HDL-C showed nominal significance (pint = 0.013), but without clear sex differences and with a greater magnitude for large HDL-P. Our work provides additional insights into a known gene-PA interaction while illustrating the importance of phenotype and model refinement toward understanding and replicating GxEs.
Collapse
Grants
- 75N92020D00005 NHLBI NIH HHS
- U54 HG003067 NHGRI NIH HHS
- UL1 TR001079 NCATS NIH HHS
- R01 HL043851 NHLBI NIH HHS
- N01HC95164 NHLBI NIH HHS
- P30 DK063491 NIDDK NIH HHS
- N01HC95165 NHLBI NIH HHS
- N01HC95159 NHLBI NIH HHS
- 75N92020D00007 NHLBI NIH HHS
- K24 HL136852 NHLBI NIH HHS
- UL1 TR000040 NCATS NIH HHS
- 75N92020D00002 NHLBI NIH HHS
- HHSN268201500003C NHLBI NIH HHS
- N01HC95160 NHLBI NIH HHS
- R01 HL160799 NHLBI NIH HHS
- R01 HL120393 NHLBI NIH HHS
- K01 DK133637 NIDDK NIH HHS
- N01HC95163 NHLBI NIH HHS
- 75N92020D00001 NHLBI NIH HHS
- N01HC95169 NHLBI NIH HHS
- N01HC95162 NHLBI NIH HHS
- 75N92020D00003 NHLBI NIH HHS
- R01 HL105756 NHLBI NIH HHS
- N01HC95168 NHLBI NIH HHS
- R01 HL118305 NHLBI NIH HHS
- UL1 TR001420 NCATS NIH HHS
- 75N92020D00004 NHLBI NIH HHS
- UM1 CA182913 NCI NIH HHS
- R01 HL156991 NHLBI NIH HHS
- N01HC95161 NHLBI NIH HHS
- R01 CA047988 NCI NIH HHS
- HHSN268201500003I NHLBI NIH HHS
- R01 HL080467 NHLBI NIH HHS
- N01HC95167 NHLBI NIH HHS
- R01 HL117626 NHLBI NIH HHS
- 75N92020D00006 NHLBI NIH HHS
- N01HC95166 NHLBI NIH HHS
- This investigation was supported by two grants from the U.S. National Heart, Lung, and Blood Institute (NHLBI), the National Institutes of Health, R01HL118305 and R01HL156991. K.E.W. was supported by K01DK133637. T.O.K. was supported by the Novo Nordisk Foundation (NNF18CC0034900, NNF21SA0072102). A.R.B. was supported by the Intramural Research Program of the National Human Genome Research Institute of the National Institutes of Health through the Center for Research on Genomics and Global Health (CRGGH). S.M. was supported by HL160799, HL117861, and K24HL136852. The WGHS is supported by the National Heart, Lung, and Blood Institute (HL043851 and HL080467) and the National Cancer Institute (CA047988 and UM1CA182913), with funding for genotyping provided by Amgen and funding for NMR assays by the American Heart Association.
- R01 HL117861 NHLBI NIH HHS
- UL1 TR001881 NCATS NIH HHS
Collapse
Affiliation(s)
- Kenneth E. Westerman
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Tuomas O. Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Magdalena Sevilla-Gonzalez
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Alexis C. Wood
- USDA/ARS Children’s Nutrition Center, Baylor College of Medicine, Houston, TX, USA
| | - Michael Y. Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Kent D. Taylor
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - James D. Otvos
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Amy R. Bentley
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Samia Mora
- Center for Lipid Metabolomics, Brigham and Women’s Hospital, Boston, MA, USA
- Division of Preventive Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Cardiovascular Division, Brigham and Women’s Hospital, Boston, MA, USA
| | - Hugues Aschard
- Department of Computational Biology, Institut Pasteur, Université de Paris, Paris, FR
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - DC Rao
- Division of Biostatistics, Washington University, St. Louis, MO, USA
| | - Charles Gu
- Division of Biostatistics, Washington University, St. Louis, MO, USA
| | - Daniel I. Chasman
- Division of Preventive Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Division of Genetics, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Alisa K. Manning
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | |
Collapse
|
2
|
Reed JN, Hasan F, Karkar A, Banka D, Hinkle J, Shastri P, Srivastava N, Scherping SC, Newkirk SE, Ferris HA, Kundu BK, Kranz S, Civelek M, Keller SR. Combined effects of genetic background and diet on mouse metabolism and gene expression. iScience 2024; 27:111323. [PMID: 39640571 PMCID: PMC11617257 DOI: 10.1016/j.isci.2024.111323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/17/2024] [Accepted: 10/30/2024] [Indexed: 12/07/2024] Open
Abstract
In humans, dietary patterns impact weight and metabolism differentially across individuals. To uncover genetic determinants for differential dietary effects, we subjected four genetically diverse mouse strains to humanized diets (American, Mediterranean, vegetarian, and vegan) with similar macronutrient composition, and performed body weight, metabolic parameter, and RNA-seq analysis. We observed pronounced diet- and strain-dependent effects on weight, and triglyceride and insulin levels. Differences in fat mass, adipose tissue, and skeletal muscle glucose uptake, and gene expression changes in most tissues were strain-dependent. In visceral adipose tissue, ∼400 genes responded to diet in a strain-dependent manner, many of them in metabolite transport and lipid metabolism pathways and several previously identified to modify diet effects in humans. Thus, genetic background profoundly impacts metabolism, though chosen dietary patterns modify the strong genetic effects. This study paves the way for future mechanistic investigations into strain-diet interactions in mice and translation to precision nutrition in humans.
Collapse
Affiliation(s)
- Jordan N. Reed
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Faten Hasan
- Department of Kinesiology, University of Virginia School of Education and Human Development, Charlottesville, VA 22903, USA
| | - Abhishek Karkar
- Department of Medicine-Division of Endocrinology and Metabolism, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Dhanush Banka
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Jameson Hinkle
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Preeti Shastri
- Department of Medicine-Division of Endocrinology and Metabolism, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Navya Srivastava
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
- Department of Radiology and Medical Imaging, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Steven C. Scherping
- Department of Medicine-Division of Endocrinology and Metabolism, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Sarah E. Newkirk
- Department of Medicine-Division of Endocrinology and Metabolism, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Heather A. Ferris
- Department of Medicine-Division of Endocrinology and Metabolism, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Bijoy K. Kundu
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
- Department of Radiology and Medical Imaging, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Sibylle Kranz
- Department of Kinesiology, University of Virginia School of Education and Human Development, Charlottesville, VA 22903, USA
| | - Mete Civelek
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Susanna R. Keller
- Department of Medicine-Division of Endocrinology and Metabolism, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| |
Collapse
|
3
|
Sun Y, Xu H, Ye K. Genome-wide association studies and multi-omics integrative analysis reveal novel loci and their molecular mechanisms for circulating polyunsaturated, monounsaturated, and saturated fatty acids. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.11.24317110. [PMID: 39606376 PMCID: PMC11601680 DOI: 10.1101/2024.11.11.24317110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Previous genome-wide association studies (GWAS) have identified genetic loci associated with the circulating levels of FAs, but the biological mechanisms of these genetic associations remain largely unexplored. Here, we conducted GWAS to identify additional genetic loci for 19 circulating fatty acid (FA) traits in UK Biobank participants of European ancestry (N = 239,268) and five other ancestries (N = 508 - 4,663). We leveraged the GWAS findings to characterize genetic correlations and colocalized regions among FAs, explore sex differences, examine FA loci influenced by lipoprotein metabolism, and apply statistical fine-mapping to pinpoint putative causal variants. We integrated GWAS signals with multi-omics quantitative trait loci (QTL) to reveal intermediate molecular phenotypes mediating the associations between the genetic loci and FA levels. Altogether, we identified 215 significant loci for polyunsaturated fatty acids (PUFAs)-related traits in European participants, 163 loci for monounsaturated fatty acids (MUFAs)-related traits, and 119 loci for saturated fatty acids (SFAs)-related traits, including 70, 61, and 54 novel loci, respectively. A novel locus for total FAs, the percentage of omega-6 PUFAs in total FAs, and total MUFAs (around genes GSTT1/2/2B) overlapped with QTL signals for all six molecular phenotypes examined, including gene expression, protein abundance, DNA methylation, splicing, histone modification, and chromatin accessibility. Across 19 FA traits, 65% of GWAS loci overlapped with QTL signals for at least one molecular phenotype. Our study identifies novel genetic loci for circulating FA levels and systematically uncovers their underlying molecular mechanisms.
Collapse
Affiliation(s)
- Yitang Sun
- Department of Genetics, University of Georgia, Athens, GA, USA
| | - Huifang Xu
- Department of Genetics, University of Georgia, Athens, GA, USA
| | - Kaixiong Ye
- Department of Genetics, University of Georgia, Athens, GA, USA
- Institute of Bioinformatics, University of Georgia, Athens, GA, USA
| |
Collapse
|
4
|
Wuni R, Vimaleswaran KS. Barriers in Translating Existing Nutrigenetics Insights to Precision Nutrition for Cardiometabolic Health in Ethnically Diverse Populations. Lifestyle Genom 2024; 17:122-135. [PMID: 39467522 DOI: 10.1159/000541909] [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: 07/01/2024] [Accepted: 09/27/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Cardiometabolic diseases pose a significant threat to global public health, with a substantial majority of cardiovascular disease mortality (more than three-quarters) occurring in low- and middle-income countries. There have been remarkable advances in recent years in identifying genetic variants that alter disease susceptibility by interacting with dietary factors. Despite the remarkable progress, several factors need to be considered before the translation of nutrigenetics insights to personalised and precision nutrition in ethnically diverse populations. Some of these factors include variations in genetic predispositions, cultural and lifestyle factors as well as socio-economic factors. SUMMARY This review aimed to explore the factors that need to be considered in bridging the gap between existing nutrigenetics insights and the implementation of personalised and precision nutrition across diverse ethnicities. Several factors might influence variations among individuals with regard to dietary exposures and metabolic responses, and these include genetic diversity, cultural and lifestyle factors as well as socio-economic factors. A multi-omics approach involving disciplines such as metabolomics, epigenetics, and the gut microbiome might contribute to improved understanding of the underlying mechanisms of gene-diet interactions and the implementation of precision nutrition although more research is needed to confirm the practicality and effectiveness of this approach. Conducting gene-diet interaction studies in diverse populations is essential and studies utilising large sample sizes are required as this improves the power to detect interactions with minimal effect sizes. Future studies should focus on replicating initial findings to enhance reliability and promote comparison across studies. Once findings have been replicated in independent samples, dietary intervention studies will be required to further strengthen the evidence and facilitate their application in clinical practice. KEY MESSAGES Nutrigenetics has a potential role to play in the prevention and management of cardiometabolic diseases. Conducting gene-diet interaction studies in diverse populations is essential giving the genetic diversity and variations in dietary patterns. Integrating data from disciplines such as metabolomics, epigenetics, and the gut microbiome could help in early identification of individuals at risk of cardiometabolic diseases as well as the implementation of precise dietary interventions for preventing and managing cardiometabolic diseases.
Collapse
Affiliation(s)
- Ramatu Wuni
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, UK
| | - Karani Santhanakrishnan Vimaleswaran
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, UK
- Institute for Food, Nutrition, and Health (IFNH), University of Reading, Reading, UK
| |
Collapse
|
5
|
Sun Y, McDonald T, Baur A, Xu H, Bateman NB, Shen Y, Li C, Ye K. Fish oil supplementation modifies the associations between genetically predicted and observed concentrations of blood lipids: a cross-sectional gene-diet interaction study in UK Biobank. Am J Clin Nutr 2024; 120:540-549. [PMID: 39019260 PMCID: PMC11393395 DOI: 10.1016/j.ajcnut.2024.07.009] [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: 09/07/2023] [Revised: 07/07/2024] [Accepted: 07/11/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND Dyslipidemia is a well-known risk factor for cardiovascular disease, the leading cause of mortality worldwide. Although habitual intake of fish oil is associated with cardioprotective effects through triglyceride reduction, the interactions of fish oil with the genetic predisposition to dysregulated lipids remain elusive. OBJECTIVES We examined whether fish oil supplementation modifies the association between genetically predicted and observed concentrations of total cholesterol, low-density lipoprotein (LDL) cholesterol, high-density lipoprotein (HDL) cholesterol, and triglycerides. METHODS A total of 441,985 participants with complete genetic and phenotypic data from the UK Biobank were included. Polygenic scores (PGS) of the 4 lipids were calculated in participants of diverse ancestries. For each lipid, multivariable linear regression models were used to assess if fish oil supplementation modified the association between PGS and the observed circulating concentration, with adjustment for relevant covariates. RESULTS Fish oil supplementation attenuates the associations between genetically predicted and observed circulating concentrations of total cholesterol, LDL cholesterol, and triglycerides while accentuating the corresponding association for HDL cholesterol among 424,090 participants of European ancestry. Consistent significant findings were obtained using PGS calculated based on multiple genome-wide association studies or alternative PGS methods. For triglycerides, each standard deviation (SD) increment in PGS is associated with 0.254 [95% confidence interval (CI): 0.248, 0.259] SD increase in the observed concentration among European-ancestry participants who reported fish oil usage. In contrast, a stronger association was observed in nonusers (0.267; 95% CI: 0.263, 0.270). Consistently, we showed that fish oil significantly attenuates the association between genetically predicted and observed concentrations of triglycerides in African-ancestry participants. CONCLUSIONS Fish oil supplementation attenuates the association between genetically predicted and observed circulating concentrations of total cholesterol, LDL cholesterol, and triglycerides while accentuating the corresponding association for HDL cholesterol in individuals of European ancestry. Further research is needed to understand the clinical implications of these findings.
Collapse
Affiliation(s)
- Yitang Sun
- Department of Genetics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, United States
| | - Tryggvi McDonald
- Department of Genetics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, United States
| | - Abigail Baur
- Department of Genetics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, United States
| | - Huifang Xu
- Department of Genetics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, United States
| | - Naveen Brahman Bateman
- Department of Genetics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, United States
| | - Ye Shen
- Department of Epidemiology and Biostatistics, College of Public Health, University of Georgia, Athens, GA, United States
| | - Changwei Li
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
| | - Kaixiong Ye
- Department of Genetics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, United States; Institute of Bioinformatics, University of Georgia, Athens, GA, United States.
| |
Collapse
|
6
|
Francis M, Westerman KE, Manning AK, Ye K. Gene-vegetarianism interactions in calcium, estimated glomerular filtration rate, and testosterone identified in genome-wide analysis across 30 biomarkers. PLoS Genet 2024; 20:e1011288. [PMID: 38990837 PMCID: PMC11239071 DOI: 10.1371/journal.pgen.1011288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 05/03/2024] [Indexed: 07/13/2024] Open
Abstract
We examined the associations of vegetarianism with metabolic biomarkers using traditional and genetic epidemiology. First, we addressed inconsistencies in self-reported vegetarianism among UK Biobank participants by utilizing data from two dietary surveys to find a cohort of strict European vegetarians (N = 2,312). Vegetarians were matched 1:4 with nonvegetarians for non-genetic association analyses, revealing significant effects of vegetarianism in 15 of 30 biomarkers. Cholesterol measures plus vitamin D were significantly lower in vegetarians, while triglycerides were higher. A genome-wide association study revealed no genome-wide significant (GWS; 5×10-8) associations with vegetarian behavior. We performed genome-wide gene-vegetarianism interaction analyses for the biomarkers, and detected a GWS interaction impacting calcium at rs72952628 (P = 4.47×10-8). rs72952628 is in MMAA, a B12 metabolic pathway gene; B12 has major deficiency potential in vegetarians. Gene-based interaction tests revealed two significant genes, RNF168 in testosterone (P = 1.45×10-6) and DOCK4 in estimated glomerular filtration rate (eGFR) (P = 6.76×10-7), which have previously been associated with testicular and renal traits, respectively. These nutrigenetic findings indicate genotype can modify the associations between vegetarianism and health outcomes.
Collapse
Affiliation(s)
- Michael Francis
- Institute of Bioinformatics, University of Georgia, Athens, Georgia, United States of America
| | - Kenneth E. Westerman
- Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, Massachusetts, United States of America
| | - Alisa K. Manning
- Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, Massachusetts, United States of America
| | - Kaixiong Ye
- Institute of Bioinformatics, University of Georgia, Athens, Georgia, United States of America
- Department of Genetics, University of Georgia, Athens, Georgia, United States of America
| |
Collapse
|
7
|
Wang J, Mou X, Lu H, Jiang H, Xian Y, Wei X, Huang Z, Tang S, Cen H, Dong M, Liang Y, Shi G. Exploring a novel seven-gene marker and mitochondrial gene TMEM38A for predicting cervical cancer radiotherapy sensitivity using machine learning algorithms. Front Endocrinol (Lausanne) 2024; 14:1302074. [PMID: 38327905 PMCID: PMC10847243 DOI: 10.3389/fendo.2023.1302074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/07/2023] [Indexed: 02/09/2024] Open
Abstract
Background Radiotherapy plays a crucial role in the management of Cervical cancer (CC), as the development of resistance by cancer cells to radiotherapeutic interventions is a significant factor contributing to treatment failure in patients. However, the specific mechanisms that contribute to this resistance remain unclear. Currently, molecular targeted therapy, including mitochondrial genes, has emerged as a new approach in treating different types of cancers, gaining significant attention as an area of research in addressing the challenge of radiotherapy resistance in cancer. Methods The present study employed a rigorous screening methodology within the TCGA database to identify a cohort of patients diagnosed with CC who had received radiotherapy treatment. The control group consisted of individuals who demonstrated disease stability or progression after undergoing radiotherapy. In contrast, the treatment group consisted of patients who experienced complete or partial remission following radiotherapy. Following this, we identified and examined the differentially expressed genes (DEGs) in the two cohorts. Subsequently, we conducted additional analyses to refine the set of excluded DEGs by employing the least absolute shrinkage and selection operator regression and random forest techniques. Additionally, a comprehensive analysis was conducted in order to evaluate the potential correlation between the expression of core genes and the extent of immune cell infiltration in patients diagnosed with CC. The mitochondrial-associated genes were obtained from the MITOCARTA 3.0. Finally, the verification of increased expression of the mitochondrial gene TMEM38A in individuals with CC exhibiting sensitivity to radiotherapy was conducted using reverse transcription quantitative polymerase chain reaction and immunohistochemistry assays. Results This process ultimately led to the identification of 7 crucial genes, viz., GJA3, TMEM38A, ID4, CDHR1, SLC10A4, KCNG1, and HMGCS2, which were strongly associated with radiotherapy sensitivity. The enrichment analysis has unveiled a significant association between these 7 crucial genes and prominent signaling pathways, such as the p53 signaling pathway, KRAS signaling pathway, and PI3K/AKT/MTOR pathway. By utilizing these 7 core genes, an unsupervised clustering analysis was conducted on patients with CC, resulting in the categorization of patients into three distinct molecular subtypes. In addition, a predictive model for the sensitivity of CC radiotherapy was developed using a neural network approach, utilizing the expression levels of these 7 core genes. Moreover, the CellMiner database was utilized to predict drugs that are closely linked to these 7 core genes, which could potentially act as crucial agents in overcoming radiotherapy resistance in CC. Conclusion To summarize, the genes GJA3, TMEM38A, ID4, CDHR1, SLC10A4, KCNG1, and HMGCS2 were found to be closely correlated with the sensitivity of CC to radiotherapy. Notably, TMEM38A, a mitochondrial gene, exhibited the highest degree of correlation, indicating its potential as a crucial biomarker for the modulation of radiotherapy sensitivity in CC.
Collapse
Affiliation(s)
- Jiajia Wang
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Xue Mou
- Department of Oncology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Haishan Lu
- Clinical Pathological Diagnosis & Research Centra, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Hai Jiang
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Yuejuan Xian
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Xilin Wei
- School of Laboratory Medicine, Youjiang Medical University for Nationalities, Baise, China
| | - Ziqiang Huang
- School of Laboratory Medicine, Youjiang Medical University for Nationalities, Baise, China
| | - Senlin Tang
- School of Laboratory Medicine, Youjiang Medical University for Nationalities, Baise, China
| | - Hongsong Cen
- School of Laboratory Medicine, Youjiang Medical University for Nationalities, Baise, China
| | - Mingyou Dong
- School of Laboratory Medicine, Youjiang Medical University for Nationalities, Baise, China
| | - Yuexiu Liang
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Guiling Shi
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| |
Collapse
|
8
|
Westerman KE, Kilpeläinen TO, Sevilla-Gonzalez M, Connelly MA, Wood AC, Tsai MY, Taylor KD, Rich SS, Rotter JI, Otvos JD, Bentley AR, Mora S, Aschard H, Rao DC, Gu C, Chasman DI, Manning AK. Refinement of a published gene-physical activity interaction impacting HDL-cholesterol: role of sex and lipoprotein subfractions. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.23.24301689. [PMID: 38313294 PMCID: PMC10836120 DOI: 10.1101/2024.01.23.24301689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
Large-scale gene-environment interaction (GxE) discovery efforts often involve compromises in the definition of outcomes and choice of covariates for the sake of data harmonization and statistical power. Consequently, refinement of exposures, covariates, outcomes, and population subsets may be helpful to establish often-elusive replication and evaluate potential clinical utility. Here, we used additional datasets, an expanded set of statistical models, and interrogation of lipoprotein metabolism via nuclear magnetic resonance (NMR)-based lipoprotein subfractions to refine a previously discovered GxE modifying the relationship between physical activity (PA) and HDL-cholesterol (HDL-C). This GxE was originally identified by Kilpeläinen et al., with the strongest cohort-specific signal coming from the Women's Genome Health Study (WGHS). We thus explored this GxE further in the WGHS (N = 23,294), with follow-up in the UK Biobank (UKB; N = 281,380), and the Multi-Ethnic Study of Atherosclerosis (MESA; N = 4,587). Self-reported PA (MET-hrs/wk), genotypes at rs295849 (nearest gene: LHX1), and NMR metabolomics data were available in all three cohorts. As originally reported, minor allele carriers of rs295849 in WGHS had a stronger positive association between PA and HDL-C (pint = 0.002). When testing a range of NMR metabolites (primarily lipoprotein and lipid subfractions) to refine the HDL-C outcome, we found a stronger interaction effect on medium-sized HDL particle concentrations (M-HDL-P; pint = 1.0×10-4) than HDL-C. Meta-regression revealed a systematically larger interaction effect in cohorts from the original meta-analysis with a greater fraction of women (p = 0.018). In the UKB, GxE effects were stronger both in women and using M-HDL-P as the outcome. In MESA, the primary interaction for HDL-C showed nominal significance (pint = 0.013), but without clear differences by sex and with a greater magnitude using large, rather than medium, HDL-P as an outcome. Towards reconciling these observations, further exploration leveraging NMR platform-specific HDL subfraction diameter annotations revealed modest agreement across all cohorts in the interaction affecting medium-to-large particles. Taken together, our work provides additional insights into a specific known gene-PA interaction while illustrating the importance of phenotype and model refinement towards understanding and replicating GxEs.
Collapse
Affiliation(s)
- Kenneth E. Westerman
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Tuomas O. Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Magdalena Sevilla-Gonzalez
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Alexis C. Wood
- USDA/ARS Children’s Nutrition Center, Baylor College of Medicine, Houston, TX, USA
| | - Michael Y. Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Kent D. Taylor
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - James D. Otvos
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Amy R. Bentley
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Samia Mora
- Center for Lipid Metabolomics, Brigham and Women’s Hospital, Boston, MA, USA
- Division of Preventive Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Cardiovascular Division, Brigham and Women’s Hospital, Boston, MA, USA
| | - Hugues Aschard
- Department of Computational Biology, Institut Pasteur, Université de Paris, Paris, FR
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - DC Rao
- Division of Biostatistics, Washington University, St. Louis, MO, USA
| | - Charles Gu
- Division of Biostatistics, Washington University, St. Louis, MO, USA
| | - Daniel I. Chasman
- Division of Preventive Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Division of Genetics, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Alisa K. Manning
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
9
|
Laguzzi F, Åkesson A, Marklund M, Qian F, Gigante B, Bartz TM, Bassett JK, Birukov A, Campos H, Hirakawa Y, Imamura F, Jäger S, Lankinen M, Murphy RA, Senn M, Tanaka T, Tintle N, Virtanen JK, Yamagishi K, Allison M, Brouwer IA, De Faire U, Eiriksdottir G, Ferrucci L, Forouhi NG, Geleijnse JM, Hodge AM, Kimura H, Laakso M, Risérus U, van Westing AC, Bandinelli S, Baylin A, Giles GG, Gudnason V, Iso H, Lemaitre RN, Ninomiya T, Post WS, Psaty BM, Salonen JT, Schulze MB, Tsai MY, Uusitupa M, Wareham NJ, Oh SW, Wood AC, Harris WS, Siscovick D, Mozaffarian D, Leander K. Role of Polyunsaturated Fat in Modifying Cardiovascular Risk Associated With Family History of Cardiovascular Disease: Pooled De Novo Results From 15 Observational Studies. Circulation 2024; 149:305-316. [PMID: 38047387 PMCID: PMC10798593 DOI: 10.1161/circulationaha.123.065530] [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: 05/10/2023] [Accepted: 10/25/2023] [Indexed: 12/05/2023]
Abstract
BACKGROUND It is unknown whether dietary intake of polyunsaturated fatty acids (PUFA) modifies the cardiovascular disease (CVD) risk associated with a family history of CVD. We assessed interactions between biomarkers of low PUFA intake and a family history in relation to long-term CVD risk in a large consortium. METHODS Blood and tissue PUFA data from 40 885 CVD-free adults were assessed. PUFA levels ≤25th percentile were considered to reflect low intake of linoleic, alpha-linolenic, and eicosapentaenoic/docosahexaenoic acids (EPA/DHA). Family history was defined as having ≥1 first-degree relative who experienced a CVD event. Relative risks with 95% CI of CVD were estimated using Cox regression and meta-analyzed. Interactions were assessed by analyzing product terms and calculating relative excess risk due to interaction. RESULTS After multivariable adjustments, a significant interaction between low EPA/DHA and family history was observed (product term pooled RR, 1.09 [95% CI, 1.02-1.16]; P=0.01). The pooled relative risk of CVD associated with the combined exposure to low EPA/DHA, and family history was 1.41 (95% CI, 1.30-1.54), whereas it was 1.25 (95% CI, 1.16-1.33) for family history alone and 1.06 (95% CI, 0.98-1.14) for EPA/DHA alone, compared with those with neither exposure. The relative excess risk due to interaction results indicated no interactions. CONCLUSIONS A significant interaction between biomarkers of low EPA/DHA intake, but not the other PUFA, and a family history was observed. This novel finding might suggest a need to emphasize the benefit of consuming oily fish for individuals with a family history of CVD.
Collapse
Affiliation(s)
- Federica Laguzzi
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine (F.L., A.A., U.D.F., K.L.), Karolinska Institutet, Stockholm, Sweden
| | - Agneta Åkesson
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine (F.L., A.A., U.D.F., K.L.), Karolinska Institutet, Stockholm, Sweden
| | - Matti Marklund
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (M.M., W.S.P)
- The George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, Australia (M.M.)
| | - Frank Qian
- Section of Cardiovascular Medicine, Boston Medical Center and Boston University Chobanian and Avedisian School of Medicine, MA (F.Q.)
- Department of Nutrition (F.Q.), Boston, MA
| | - Bruna Gigante
- Cardiovascular Medicine Unit, Department of Medicine Solna (B.G.), Karolinska Institutet, Stockholm, Sweden
| | - Traci M. Bartz
- Cardiovascular Health Research Unit, Departments of Biostatistics (T.M.B.), University of Washington, Seattle
- Medicine (T.M.B., R.N.L., B.M.P.), University of Washington, Seattle
| | - Julie K. Bassett
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Australia (J.K.B., A.M.H., G.G.G.)
| | - Anna Birukov
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal (A.K.B., S.J., M.B.S.)
- German Center for Diabetes Research, Neuherberg (A.K.B., S.J., M.B.S.)
| | - Hannia Campos
- Harvard T.H. Chan School of Public Health (H.C.), Boston, MA
| | - Yoichiro Hirakawa
- Departments of Epidemiology and Public Health and Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan (Y.H., T.N.)
| | - Fumiaki Imamura
- Medical Research Council Epidemiology Unit, University of Cambridge School of Clinical Medicine, UK (F.I., N.G.F., N.J.W.)
| | - Susanne Jäger
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal (A.K.B., S.J., M.B.S.)
| | - Maria Lankinen
- Institutes of Public Health and Clinical Nutrition (M. Lankinen, J.K.V., M.U.), University of Eastern Finland, Kuopio
| | - Rachel A. Murphy
- Cancer Control Research, BC Cancer Agency, Vancouver, Canada (R.A.M.)
- School of Population and Public Health, University of British Columbia, Vancouver, Canada (R.A.M.)
| | - Mackenzie Senn
- United States Department of Agriculture/Agricultural Research Service Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX (M.S., A.C.W.)
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany (M.B.S.)
| | - Toshiko Tanaka
- Longitudinal Study Section, National Institute on Aging, Baltimore, MD (T.T., L.F.)
| | - Nathan Tintle
- Fatty Acid Research Institute, Sioux Falls, SD (N.T., W.S.H.)
- Department of Population Health Nursing Science, University of Illinois – Chicago (N.T.)
| | - Jyrki K. Virtanen
- Institutes of Public Health and Clinical Nutrition (M. Lankinen, J.K.V., M.U.), University of Eastern Finland, Kuopio
| | - Kazumasa Yamagishi
- Department of Public Health Medicine, Institute of Medicine (K.Y., H.K.), University of Tsukuba, Japan
- Health Services Research and Development Center (K.Y., H.K.), University of Tsukuba, Japan
| | - Matthew Allison
- Department of Family Medicine, University of California, San Diego, La Jolla (M.A.)
| | - Ingeborg A. Brouwer
- Department of Health Sciences, Faculty of Science, Vrije Universiteit Amsterdam, The Netherlands (I.A.B.)
- Amsterdam Public Health Research Institute, The Netherlands (I.A.B.)
| | - Ulf De Faire
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine (F.L., A.A., U.D.F., K.L.), Karolinska Institutet, Stockholm, Sweden
| | | | - Luigi Ferrucci
- Longitudinal Study Section, National Institute on Aging, Baltimore, MD (T.T., L.F.)
| | - Nita G. Forouhi
- Medical Research Council Epidemiology Unit, University of Cambridge School of Clinical Medicine, UK (F.I., N.G.F., N.J.W.)
| | - Johanna M. Geleijnse
- Division of Human Nutrition and Health, Wageningen University and Research, The Netherlands (J.M.G., A.C.v.W.)
| | - Allison M Hodge
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Australia (J.K.B., A.M.H., G.G.G.)
- Centre for Epidemiology and Biostatistics, University of Melbourne, Victoria, Australia (A.M.H., G.G.G.)
| | - Hitomi Kimura
- Department of Public Health Medicine, Institute of Medicine (K.Y., H.K.), University of Tsukuba, Japan
- Health Services Research and Development Center (K.Y., H.K.), University of Tsukuba, Japan
| | - Markku Laakso
- Clinical Medicine, Internal Medicine (M. Laakso), University of Eastern Finland, Kuopio
- Kuopio University Hospital (M. Laakso), University of Eastern Finland, Kuopio
| | - Ulf Risérus
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Sweden (M.M., U.R)
| | - Anniek C. van Westing
- United States Department of Agriculture/Agricultural Research Service Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX (M.S., A.C.W.)
- Division of Human Nutrition and Health, Wageningen University and Research, The Netherlands (J.M.G., A.C.v.W.)
| | - Stefania Bandinelli
- Geriatric Unit, Azienda Unità Sanitaria Locale Toscana Centro, Florence, Italy (S.B.)
| | - Ana Baylin
- University of Michigan School of Public Health, Ann Arbor (A. Baylin)
| | - Graham G. Giles
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Australia (J.K.B., A.M.H., G.G.G.)
- Centre for Epidemiology and Biostatistics, University of Melbourne, Victoria, Australia (A.M.H., G.G.G.)
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Victoria, Australia (G.G.G.)
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur (G.E., V.G.)
- Faculty of Medicine, University of Iceland, Reykjavik (V.G.)
| | - Hiroyasu Iso
- Public Health, Department of Social Medicine, Osaka University Graduate School of Medicine, Suita, Japan (H.I.)
- Institute for Global Health Policy Research, Bureau of International Health Cooperation, National Center for Global Health and Medicine, Tokyo, Japan (H.I.)
| | | | - Toshiharu Ninomiya
- Departments of Epidemiology and Public Health and Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan (Y.H., T.N.)
| | - Wendy S. Post
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (M.M., W.S.P)
- Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD (W.S.P.)
| | - Bruce M. Psaty
- Medicine (T.M.B., R.N.L., B.M.P.), University of Washington, Seattle
- Epidemiology (B.M.P.), University of Washington, Seattle
- Health Systems and Population Health (B.M.P.), University of Washington, Seattle
| | - Jukka T. Salonen
- Metabolic Analytical Services Oy, Helsinki, Finland (J.T.S.)
- University of Helsinki, the Faculty of Medicine, Department of Public Health, Finland (J.T.S.)
| | - Matthias B. Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal (A.K.B., S.J., M.B.S.)
- German Center for Diabetes Research, Neuherberg (A.K.B., S.J., M.B.S.)
| | - Michael Y. Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis (M.Y.T.)
| | - Matti Uusitupa
- Institutes of Public Health and Clinical Nutrition (M. Lankinen, J.K.V., M.U.), University of Eastern Finland, Kuopio
| | - Nicholas J. Wareham
- Medical Research Council Epidemiology Unit, University of Cambridge School of Clinical Medicine, UK (F.I., N.G.F., N.J.W.)
| | - Seung-Won Oh
- Department of Family Medicine, Seoul National University College of Medicine, and Healthcare System Gangnam Center, Seoul National University Hospital, Republic of Korea (S.W.O.)
| | - Alexis C. Wood
- United States Department of Agriculture/Agricultural Research Service Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX (M.S., A.C.W.)
| | - William S. Harris
- Fatty Acid Research Institute, Sioux Falls, SD (N.T., W.S.H.)
- Department of Internal Medicine, Sanford School of Medicine, University of South Dakota, Sioux Falls (W.S.H.)
| | | | - Dariush Mozaffarian
- Food Is Medicine Institute, Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA (D.M.)
| | - Karin Leander
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine (F.L., A.A., U.D.F., K.L.), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
10
|
Erbay MI, Gamarra Valverde NN, Patel P, Ozkan HS, Wilson A, Banerjee S, Babazade A, Londono V, Sood A, Gupta R. Fish Oil Derivatives in Hypertriglyceridemia: Mechanism and Cardiovascular Prevention: What Do Studies Say? Curr Probl Cardiol 2024; 49:102066. [PMID: 37657524 DOI: 10.1016/j.cpcardiol.2023.102066] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 08/26/2023] [Indexed: 09/03/2023]
Abstract
Hypertriglyceridemia is a type of dyslipidemia characterized by high triglyceride levels in the blood and increases the risk of cardiovascular disease. Conventional management includes antilipidemic medications such as statins, lowering LDL and triglyceride levels as well as raising HDL levels. However, the treatment may be stratified using omega-3 fatty acid supplements such as eicosatetraenoic acid (EPA) and docosahexaenoic acid (DHA), aka fish oil derivatives. Studies have shown that fish oil supplements reduce the risk of cardiovascular diseases; however, the underlying mechanism and the extent of reduction in CVD need more clarification. Our paper aims to review the clinical trials and observational studies in the current literature, investigating the use of fish oil and its benefits on the cardiovascular system as well as the proposed underlying mechanism.
Collapse
Affiliation(s)
- Muhammed Ibrahim Erbay
- Department of Medicine, Istanbul University Cerrahpasa, Cerrahpasa School of Medicine, Istanbul, Turkey
| | - Norma Nicole Gamarra Valverde
- Department of Medicine, Alberto Hurtado Faculty of Human Medicine, Facultad de Medicina Alberto Hurtado, Universidad Peruana Cayetano Heredia, Lima, Perú
| | - Parth Patel
- Department of Medicine, University of Missouri Kansas City School of Medicine, Kansas City, MI
| | - Hasan Selcuk Ozkan
- Department of Medicine, Ege University, School of Medicine, Izmir, Turkey
| | - Andre Wilson
- Department of Medicine, Howard University College of Medicine, Washington, D.C
| | - Suvam Banerjee
- Department of Health and Family Welfare, Burdwan Medical College and Hospital, The West Bengal University of Health Sciences, Government of West Bengal, India
| | - Aydan Babazade
- Department of Medicine, Azerbaijan Medical University, School of Medicine, Baku, Azerbaijan
| | - Valeria Londono
- Department of Medicine, Georgetown University School of Medicine, Washington, D.C
| | - Aayushi Sood
- Department of Internal Medicine, The Wright Center for Graduate Medical Education, Scranton, PA
| | - Rahul Gupta
- Department of Cardiology, Lehigh Valley Health Network, Allentown, PA.
| |
Collapse
|
11
|
Sun Y, McDonald T, Baur A, Xu H, Bateman NB, Shen Y, Li C, Ye K. Fish oil supplementation modifies the genetic potential for blood lipids. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.22.23295987. [PMID: 37808791 PMCID: PMC10557817 DOI: 10.1101/2023.09.22.23295987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Background Dyslipidemia is a well-known risk factor for cardiovascular disease, which has been the leading cause of mortality worldwide. Although habitual intake of fish oil has been implicated in offering cardioprotective effects through triglyceride reduction, the interactions of fish oil with the genetic predisposition to dysregulated lipids remain elusive. Objectives We examined whether fish oil supplementation can modify the genetic potential for the circulating levels of four lipids, including total cholesterol, low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), and triglycerides. Methods A total of 441,985 participants with complete genetic and phenotypic data from the UK Biobank were included in our study. Polygenic scores (PGS) were calculated in participants of diverse ancestries. Multivariable linear regression models were used to assess associations with adjustment for relevant risk factors. Results Fish oil supplementation mitigated genetic susceptibility to elevated levels of total cholesterol, LDL-C, and triglycerides, while amplifying genetic potential for increased HDL-C among 424,090 participants of European ancestry P interaction < 0.05 . Consistent significant findings were obtained using PGS calculated based on multiple genome-wide association studies or alternative PGS methods. We also showed that fish oil significantly attenuated genetic predisposition to high triglycerides in African-ancestry participants. Conclusions Fish oil supplementation attenuated the genetic susceptibility to elevated blood levels of total cholesterol, LDL-C, and triglycerides, while accentuating genetic potential for higher HDL-C. These results suggest that fish oil may have a beneficial impact on modifying genome-wide genetic effects on elevated lipid levels in the general population.
Collapse
Affiliation(s)
- Yitang Sun
- Department of Genetics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA
| | - Tryggvi McDonald
- Department of Genetics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA
| | - Abigail Baur
- Department of Genetics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA
| | - Huifang Xu
- Department of Genetics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA
| | - Naveen Brahman Bateman
- Department of Genetics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA
| | - Ye Shen
- Department of Epidemiology and Biostatistics, College of Public Health, University of Georgia, Athens, GA, USA
| | - Changwei Li
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Kaixiong Ye
- Department of Genetics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA
- Institute of Bioinformatics, University of Georgia, Athens, GA, USA
| |
Collapse
|
12
|
Snaebjarnarson AS, Helgadottir A, Arnadottir GA, Ivarsdottir EV, Thorleifsson G, Ferkingstad E, Einarsson G, Sveinbjornsson G, Thorgeirsson TE, Ulfarsson MO, Halldorsson BV, Olafsson I, Erikstrup C, Pedersen OB, Nyegaard M, Bruun MT, Ullum H, Brunak S, Iversen KK, Christensen AH, Olesen MS, Ghouse J, Banasik K, Knowlton KU, Arnar DO, Thorgeirsson G, Nadauld L, Ostrowski SR, Bundgaard H, Holm H, Sulem P, Stefansson K, Gudbjartsson DF. Complex effects of sequence variants on lipid levels and coronary artery disease. Cell 2023; 186:4085-4099.e15. [PMID: 37714134 DOI: 10.1016/j.cell.2023.08.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 05/06/2023] [Accepted: 08/10/2023] [Indexed: 09/17/2023]
Abstract
Many sequence variants have additive effects on blood lipid levels and, through that, on the risk of coronary artery disease (CAD). We show that variants also have non-additive effects and interact to affect lipid levels as well as affecting variance and correlations. Variance and correlation effects are often signatures of epistasis or gene-environmental interactions. These complex effects can translate into CAD risk. For example, Trp154Ter in FUT2 protects against CAD among subjects with the A1 blood group, whereas it associates with greater risk of CAD in others. His48Arg in ADH1B interacts with alcohol consumption to affect lipid levels and CAD. The effect of variants in TM6SF2 on blood lipids is greatest among those who never eat oily fish but absent from those who often do. This work demonstrates that variants that affect variance of quantitative traits can allow for the discovery of epistasis and interactions of variants with the environment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Magnus O Ulfarsson
- deCODE genetics/Amgen, Inc., Reykjavik 102, Iceland; Faculty of Electrical and Computer Engineering, University of Iceland, Reykjavik 102, Iceland
| | | | - Isleifur Olafsson
- Department of Clinical Biochemistry, Landspitali - National University Hospital of Iceland, Hringbraut, Reykjavik 101, Iceland
| | - Christian Erikstrup
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus 8200, Denmark; Department of Clinical Medicine, Health, Aarhus University, Aarhus 8200, Denmark
| | - Ole B Pedersen
- Department of Clinical Immunology, Zealand University Hospital, Køge 4600, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen 1165, Denmark
| | - Mette Nyegaard
- Department of Health Science and Technology, Faculty of Medicine, Aalborg University, Aalborg 9220, Denmark
| | - Mie T Bruun
- Department of Clinical Immunology, Odense University Hospital, Odense 5000, Denmark
| | - Henrik Ullum
- Statens Serum Institut, Copenhagen 2300, Denmark
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Kasper Karmark Iversen
- Department of Clinical Medicine, University of Copenhagen, Copenhagen 1165, Denmark; Department of Emergency Medicine, Copenhagen University Hospital Herlev and Gentofte, Herlev 2900, Denmark; Department of Cardiology, Copenhagen University Hospital, Herlev-Gentofte Hospital, Herlev 2900, Denmark
| | - Alex Hoerby Christensen
- Department of Clinical Medicine, University of Copenhagen, Copenhagen 1165, Denmark; Department of Cardiology, Copenhagen University Hospital, Herlev-Gentofte Hospital, Herlev 2900, Denmark
| | - Morten S Olesen
- Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen 2100, Denmark; Laboratory for Molecular Cardiology, Department of Biomedical Sciences, University of Copenhagen, Copenhagen 1165, Denmark
| | - Jonas Ghouse
- Laboratory for Molecular Cardiology, Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen 2100, Denmark; Laboratory for Molecular Cardiology, Department of Biomedical Sciences, University of Copenhagen, Copenhagen 1165, Denmark
| | - Karina Banasik
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Kirk U Knowlton
- Intermountain Medical Center, Intermountain Heart Institute, Salt Lake City, UT 84143, USA
| | - David O Arnar
- deCODE genetics/Amgen, Inc., Reykjavik 102, Iceland; Faculty of Medicine, University of Iceland, Vatnsmyrarvegur, Reykjavik 101, Iceland; Division of Cardiology, Department of Internal Medicine, Landspitali - National University Hospital of Iceland, Hringbraut, Reykjavik 101, Iceland
| | - Gudmundur Thorgeirsson
- deCODE genetics/Amgen, Inc., Reykjavik 102, Iceland; Faculty of Medicine, University of Iceland, Vatnsmyrarvegur, Reykjavik 101, Iceland; Division of Cardiology, Department of Internal Medicine, Landspitali - National University Hospital of Iceland, Hringbraut, Reykjavik 101, Iceland
| | - Lincoln Nadauld
- Precision Genomics, Intermountain Healthcare, Saint George, UT 84790, USA
| | - Sisse Rye Ostrowski
- Department of Clinical Medicine, University of Copenhagen, Copenhagen 1165, Denmark; Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen 2100, Denmark
| | - Henning Bundgaard
- Department of Clinical Medicine, University of Copenhagen, Copenhagen 1165, Denmark; Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen 2100, Denmark
| | - Hilma Holm
- deCODE genetics/Amgen, Inc., Reykjavik 102, Iceland
| | | | - Kari Stefansson
- deCODE genetics/Amgen, Inc., Reykjavik 102, Iceland; Faculty of Medicine, University of Iceland, Vatnsmyrarvegur, Reykjavik 101, Iceland.
| | - Daniel F Gudbjartsson
- deCODE genetics/Amgen, Inc., Reykjavik 102, Iceland; School of Engineering and Natural Sciences, University of Iceland, Reykjavik 102, Iceland.
| |
Collapse
|
13
|
Shimizu N, Shiraishi H, Hanada T. Zebrafish as a Useful Model System for Human Liver Disease. Cells 2023; 12:2246. [PMID: 37759472 PMCID: PMC10526867 DOI: 10.3390/cells12182246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/31/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Liver diseases represent a significant global health challenge, thereby necessitating extensive research to understand their intricate complexities and to develop effective treatments. In this context, zebrafish (Danio rerio) have emerged as a valuable model organism for studying various aspects of liver disease. The zebrafish liver has striking similarities to the human liver in terms of structure, function, and regenerative capacity. Researchers have successfully induced liver damage in zebrafish using chemical toxins, genetic manipulation, and other methods, thereby allowing the study of disease mechanisms and the progression of liver disease. Zebrafish embryos or larvae, with their transparency and rapid development, provide a unique opportunity for high-throughput drug screening and the identification of potential therapeutics. This review highlights how research on zebrafish has provided valuable insights into the pathological mechanisms of human liver disease.
Collapse
Affiliation(s)
- Nobuyuki Shimizu
- Department of Cell Biology, Oita University Faculty of Medicine, Yufu 879-5593, Oita, Japan;
| | | | - Toshikatsu Hanada
- Department of Cell Biology, Oita University Faculty of Medicine, Yufu 879-5593, Oita, Japan;
| |
Collapse
|
14
|
Westerman KE, Walker ME, Gaynor SM, Wessel J, DiCorpo D, Ma J, Alonso A, Aslibekyan S, Baldridge AS, Bertoni AG, Biggs ML, Brody JA, Chen YDI, Dupuis J, Goodarzi MO, Guo X, Hasbani NR, Heath A, Hidalgo B, Irvin MR, Johnson WC, Kalyani RR, Lange L, Lemaitre RN, Liu CT, Liu S, Moon JY, Nassir R, Pankow JS, Pettinger M, Raffield LM, Rasmussen-Torvik LJ, Selvin E, Senn MK, Shadyab AH, Smith AV, Smith NL, Steffen L, Talegakwar S, Taylor KD, de Vries PS, Wilson JG, Wood AC, Yanek LR, Yao J, Zheng Y, Boerwinkle E, Morrison AC, Fornage M, Russell TP, Psaty BM, Levy D, Heard-Costa NL, Ramachandran VS, Mathias RA, Arnett DK, Kaplan R, North KE, Correa A, Carson A, Rotter JI, Rich SS, Manson JE, Reiner AP, Kooperberg C, Florez JC, Meigs JB, Merino J, Tobias DK, Chen H, Manning AK. Investigating Gene-Diet Interactions Impacting the Association Between Macronutrient Intake and Glycemic Traits. Diabetes 2023; 72:653-665. [PMID: 36791419 PMCID: PMC10130485 DOI: 10.2337/db22-0851] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 02/02/2023] [Indexed: 02/17/2023]
Abstract
Few studies have demonstrated reproducible gene-diet interactions (GDIs) impacting metabolic disease risk factors, likely due in part to measurement error in dietary intake estimation and insufficient capture of rare genetic variation. We aimed to identify GDIs across the genetic frequency spectrum impacting the macronutrient-glycemia relationship in genetically and culturally diverse cohorts. We analyzed 33,187 participants free of diabetes from 10 National Heart, Lung, and Blood Institute Trans-Omics for Precision Medicine program cohorts with whole-genome sequencing, self-reported diet, and glycemic trait data. We fit cohort-specific, multivariable-adjusted linear mixed models for the effect of diet, modeled as an isocaloric substitution of carbohydrate for fat, and its interactions with common and rare variants genome-wide. In main effect meta-analyses, participants consuming more carbohydrate had modestly lower glycemic trait values (e.g., for glycated hemoglobin [HbA1c], -0.013% HbA1c/250 kcal substitution). In GDI meta-analyses, a common African ancestry-enriched variant (rs79762542) reached study-wide significance and replicated in the UK Biobank cohort, indicating a negative carbohydrate-HbA1c association among major allele homozygotes only. Simulations revealed that >150,000 samples may be necessary to identify similar macronutrient GDIs under realistic assumptions about effect size and measurement error. These results generate hypotheses for further exploration of modifiable metabolic disease risk in additional cohorts with African ancestry. ARTICLE HIGHLIGHTS We aimed to identify genetic modifiers of the dietary macronutrient-glycemia relationship using whole-genome sequence data from 10 Trans-Omics for Precision Medicine program cohorts. Substitution models indicated a modest reduction in glycemia associated with an increase in dietary carbohydrate at the expense of fat. Genome-wide interaction analysis identified one African ancestry-enriched variant near the FRAS1 gene that may interact with macronutrient intake to influence hemoglobin A1c. Simulation-based power calculations accounting for measurement error suggested that substantially larger sample sizes may be necessary to discover further gene-macronutrient interactions.
Collapse
Affiliation(s)
- Kenneth E. Westerman
- Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, MA
| | - Maura E. Walker
- Department of Medicine, Section of Preventive Medicine, Boston University School of Medicine, Boston, MA
- Department of Health Sciences, Sargent College of Health and Rehabilitation Sciences, Boston University, Boston, MA
| | - Sheila M. Gaynor
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Jennifer Wessel
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indianapolis, IN
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
- Diabetes Translational Research Center, Indiana University, Indianapolis, IN
| | - Daniel DiCorpo
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Jiantao Ma
- Nutrition Epidemiology and Data Science, Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA
| | - Alvaro Alonso
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | | | - Abigail S. Baldridge
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Alain G. Bertoni
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC
| | - Mary L. Biggs
- Department of Biostatistics, University of Washington, Seattle, WA
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA
| | - Jennifer A. Brody
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Joseé Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Mark O. Goodarzi
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Natalie R. Hasbani
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Adam Heath
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Bertha Hidalgo
- School of Public Health, University of Alabama at Birmingham, Birmingham, AL
| | - Marguerite R. Irvin
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL
| | - W. Craig Johnson
- Department of Biostatistics, University of Washington, Seattle, WA
| | - Rita R. Kalyani
- GeneSTAR Research Program, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Leslie Lange
- Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO
| | - Rozenn N. Lemaitre
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA
- Department of Internal Medicine, University of Washington, Seattle, WA
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
- National Heart, Lung, and Blood Institute and Boston University’s Framingham Heart Study, Framingham, MA
- Evans Department of Medicine, Section of Preventive Medicine and Epidemiology, Boston University School of Medicine, Boston, MA
- Evans Department of Medicine, Whitaker Cardiovascular Institute and Cardiology Section, Boston University School of Medicine, Boston, MA
| | - Simin Liu
- Center for Global Cardiometabolic Health, Boston, MA
| | - Jee-Young Moon
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Rami Nassir
- Department of Pathology, School of Medicine, Umm Al-Qura University, Mecca, Saudi Arabia
| | - James S. Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Mary Pettinger
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Laura M. Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | | | - Elizabeth Selvin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Mackenzie K. Senn
- USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX
| | - Aladdin H. Shadyab
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, La Jolla, CA
| | - Albert V. Smith
- Department of Biostatistics, University of Michigan, Ann Arbor, MI
| | - Nicholas L. Smith
- Department of Epidemiology, University of Washington, Seattle, WA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA
- Department of Veterans Affairs Office of Research and Development, Seattle Epidemiologic Research and Information Center, Seattle, WA
| | - Lyn Steffen
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Sameera Talegakwar
- Department of Exercise and Nutrition Sciences, Milken Institute School of Public Health, The George Washington University, Washington, DC
| | - Kent D. Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Paul S. de Vries
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - James G. Wilson
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Alexis C. Wood
- USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX
| | - Lisa R. Yanek
- GeneSTAR Research Program, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Yinan Zheng
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX
| | - Alanna C. Morrison
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Miriam Fornage
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Tracy P. Russell
- Department of Pathology and Laboratory Medicine, University of Vermont Larner College of Medicine, Burlington, VT
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
- Department of Epidemiology, University of Washington, Seattle, WA
- Department of Health Systems and Population Health, University of Washington, Seattle, WA
| | - Daniel Levy
- National Heart, Lung, and Blood Institute and Boston University’s Framingham Heart Study, Framingham, MA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Nancy L. Heard-Costa
- National Heart, Lung, and Blood Institute and Boston University’s Framingham Heart Study, Framingham, MA
- Department of Neurology, Boston University School of Medicine, Boston, MA
| | - Vasan S. Ramachandran
- National Heart, Lung, and Blood Institute and Boston University’s Framingham Heart Study, Framingham, MA
- Evans Department of Medicine, Section of Preventive Medicine and Epidemiology, Boston University School of Medicine, Boston, MA
- Evans Department of Medicine, Whitaker Cardiovascular Institute and Cardiology Section, Boston University School of Medicine, Boston, MA
| | - Rasika A. Mathias
- GeneSTAR Research Program, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Donna K. Arnett
- College of Public Health, University of Kentucky, Lexington, KY
| | - Robert Kaplan
- Clinical Excellence Research Center, School of Medicine, Stanford University, Stanford, CA
| | - Kari E. North
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Adolfo Correa
- Department of Population Health Science, University of Mississippi Medical Center, Jackson, MS
| | - April Carson
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Stephen S. Rich
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, VA
| | | | | | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Jose C. Florez
- Department of Medicine, Harvard Medical School, Boston, MA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, MA
- Diabetes Unit, Massachusetts General Hospital, Boston, MA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
| | - James B. Meigs
- Department of Medicine, Harvard Medical School, Boston, MA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, MA
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA
| | - Jordi Merino
- Department of Medicine, Harvard Medical School, Boston, MA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, MA
- Diabetes Unit, Massachusetts General Hospital, Boston, MA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
| | - Deirdre K. Tobias
- Division of Preventive Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Han Chen
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
- Center for Precision Health, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Alisa K. Manning
- Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, MA
| |
Collapse
|
15
|
Francis M, Li C, Sun Y, Zhou J, Li X, Brenna JT, Ye K. Correction: Genome-wide association study of fish oil supplementation on lipid traits in 81,246 individuals reveals new gene-diet interaction loci. PLoS Genet 2023; 19:e1010735. [PMID: 37071599 PMCID: PMC10112772 DOI: 10.1371/journal.pgen.1010735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2023] Open
Abstract
[This corrects the article DOI: 10.1371/journal.pgen.1009431.].
Collapse
|
16
|
Update of a Genetic Risk Score Predictive of the Plasma Triglyceride Response to an Omega-3 Fatty Acid Supplementation in the FAS Study. Nutrients 2023; 15:nu15051156. [PMID: 36904157 PMCID: PMC10005670 DOI: 10.3390/nu15051156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/17/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023] Open
Abstract
A genetic risk score (GRS) predictive of the plasma triglyceride (TG) response to an omega-3 fatty acid (n-3 FA) supplementation has been previously developed in the Fatty Acid Sensor (FAS) Study. Recently, novel single nucleotide polymorphisms (SNPs) interacting with a fish oil supplementation and associated with plasma lipid levels have been identified in the UK Biobank. The aim of this study was to verify whether the addition of SNPs identified in the UK Biobank to the GRS built in the FAS Study improves its capacity to predict the plasma TG response to an n-3 FA supplementation. SNPs interacting with fish oil supplementation in the modulation of plasma lipid levels in the UK Biobank and associated with plasma TG levels have been genotyped in participants of the FAS Study (n = 141). Participants have been supplemented with 5 g fish oil/day for six weeks. Plasma TG concentrations were measured before and after the supplementation. Based on the initial GRS of 31 SNPs (GRS31), we computed three new GRSs by adding new SNPs identified in the UK Biobank: GRS32 (rs55707100), GRS38 (seven new SNPs specifically associated with plasma TG levels), and GRS46 (all 15 new SNPs associated with plasma lipid levels). The initial GRS31 explained 50.1% of the variance in plasma TG levels during the intervention, whereas GRS32, GRS38, and GRS46 explained 49.1%, 45.9%, and 45%, respectively. A significant impact on the probability of being classified as a responder or a nonresponder was found for each of the GRSs analyzed, but none of them outperformed the predictive capacity of GRS31 in any of the metrics analyzed, i.e., accuracy, area under the response operating curve (AUC-ROC), sensitivity, specificity and McFadden's pseudo R2. The addition of SNPs identified in the UK Biobank to the initial GRS31 did not significantly improve its capacity to predict the plasma TG response to an n-3 FA supplementation. Thus, GRS31 still remains the most precise tool so far by which to discriminate the individual responsiveness to n-3 FAs. Further studies are needed in the field to increase our knowledge of factors underlying the heterogeneity observed in the metabolic response to an n-3 FA supplementation.
Collapse
|
17
|
Tao J, Helali AE, Ho JC, Lam WWT, Pang H. An Evaluation of Gene-Diet Interaction Statistical Methods and Discovery of rs7175421-Whole Grain Interaction in Lung Cancer. Nutr Cancer 2022; 75:219-227. [PMID: 35930377 DOI: 10.1080/01635581.2022.2104878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Dietary factors show different effects on genetically diverse populations. Scientific research uses gene-environment interaction models to study the effects of dietary factors on genetically diverse populations for lung cancer risk. However, previous study designs have not investigated the degree of type I error inflation and, in some instances, have not corrected for multiple testing. Using a motivating investigation of diet-gene interaction and lung cancer risk, we propose a training and testing strategy and perform real-world simulations to select the appropriate statistical methods to reduce false-positive discoveries. The simulation results show that the unconstrained maximum likelihood (UML) method controls the type I error better than the constrained maximum likelihood (CML). The empirical Bayesian (EB) method can compete with the UML method in achieving statistical power and controlling type I error. We observed a significant interaction between SNP rs7175421 with dietary whole grain in lung cancer prevention, with an effect size (standard error) of -0.312 (0.112) for EB estimate. SNP rs7175421 may interact with dietary whole grains in modulating lung cancer risk. Evaluating statistical methods for gene-diet interaction analysis can help balance the statistical power and type I error.
Collapse
Affiliation(s)
- Jun Tao
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong, China
| | - Aya El Helali
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong, China
| | - James C Ho
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong, China
| | - Wendy W T Lam
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong, China.,Jockey Club Institute of Cancer Care, The University of Hong Kong, Pok Fu Lam, Hong Kong, China
| | - Herbert Pang
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong, China.,Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
18
|
Westerman KE, Majarian TD, Giulianini F, Jang DK, Miao J, Florez JC, Chen H, Chasman DI, Udler MS, Manning AK, Cole JB. Variance-quantitative trait loci enable systematic discovery of gene-environment interactions for cardiometabolic serum biomarkers. Nat Commun 2022; 13:3993. [PMID: 35810165 PMCID: PMC9271055 DOI: 10.1038/s41467-022-31625-5] [Citation(s) in RCA: 27] [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: 11/15/2021] [Accepted: 06/24/2022] [Indexed: 11/29/2022] Open
Abstract
Gene-environment interactions represent the modification of genetic effects by environmental exposures and are critical for understanding disease and informing personalized medicine. These often induce differential phenotypic variance across genotypes; these variance-quantitative trait loci can be prioritized in a two-stage interaction detection strategy to greatly reduce the computational and statistical burden and enable testing of a broader range of exposures. We perform genome-wide variance-quantitative trait locus analysis for 20 serum cardiometabolic biomarkers by multi-ancestry meta-analysis of 350,016 unrelated participants in the UK Biobank, identifying 182 independent locus-biomarker pairs (p < 4.5×10-9). Most are concentrated in a small subset (4%) of loci with genome-wide significant main effects, and 44% replicate (p < 0.05) in the Women's Genome Health Study (N = 23,294). Next, we test each locus-biomarker pair for interaction across 2380 exposures, identifying 847 significant interactions (p < 2.4×10-7), of which 132 are independent (p < 0.05) after accounting for correlation between exposures. Specific examples demonstrate interaction of triglyceride-associated variants with distinct body mass- versus body fat-related exposures as well as genotype-specific associations between alcohol consumption and liver stress at the ADH1B gene. Our catalog of variance-quantitative trait loci and gene-environment interactions is publicly available in an online portal.
Collapse
Affiliation(s)
- Kenneth E Westerman
- Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital, Boston, MA, USA.
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| | - Timothy D Majarian
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Franco Giulianini
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Dong-Keun Jang
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Jenkai Miao
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, USA
| | - Jose C Florez
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Han Chen
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Medical and Population Genetics Program, Broad Institute, Cambridge, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Miriam S Udler
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Alisa K Manning
- Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Joanne B Cole
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, USA.
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
19
|
Associations between Omega-3 Index, Dopaminergic Genetic Variants and Aggressive and Metacognitive Traits: A Study in Adult Male Prisoners. Nutrients 2022; 14:nu14071379. [PMID: 35405990 PMCID: PMC9002862 DOI: 10.3390/nu14071379] [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: 02/28/2022] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 12/22/2022] Open
Abstract
Omega-3 long-chain polyunsaturated fatty acids (n-3 LCPUFA) are critical for cell membrane structure and function. Human beings have a limited ability to synthesise docosahexaenoic acid (DHA), the main n-3 LCPUFA required for neurological development. Inadequate levels of n-3 LCPUFA can affect the dopaminergic system in the brain and, when combined with genetic and other factors, increase the risk of developing aggression, inattention and impulse-control disorders. In this study, male prisoners were administered questionnaires assessing aggressive behaviour and executive functions. Participants also produced blood sampling for the measurement of the Omega-3 Index and the genotyping of dopaminergic genetic variants. Significant associations were found between functional genetic polymorphism in DBH rs1611115 and verbal aggression and between DRD2 rs4274224 and executive functions. However, the Omega-3 Index was not significantly associated with the tested dopaminergic polymorphisms. Although previous interactions between specific genotypes and n-3 LCPUFA were previously reported, they remain limited and poorly understood. We did not find any association between n-3 LCPUFA and dopaminergic polymorphisms in adult male prisoners; however, we confirmed the importance of genetic predisposition for dopaminergic genes (DBH and DRD2) in aggressive behaviour, memory dysfunction and attention-deficit disorder.
Collapse
|
20
|
San-Cristobal R, de Toro-Martín J, Vohl MC. Appraisal of Gene-Environment Interactions in GWAS for Evidence-Based Precision Nutrition Implementation. Curr Nutr Rep 2022; 11:563-573. [PMID: 35948824 PMCID: PMC9750926 DOI: 10.1007/s13668-022-00430-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/15/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW This review aims to analyse the currently reported gene-environment (G × E) interactions in genome-wide association studies (GWAS), involving environmental factors such as lifestyle and dietary habits related to metabolic syndrome phenotypes. For this purpose, the present manuscript reviews the available GWAS registered on the GWAS Catalog reporting the interaction between environmental factors and metabolic syndrome traits. RECENT FINDINGS Advances in omics-related analytical and computational approaches in recent years have led to a better understanding of the biological processes underlying these G × E interactions. A total of 42 GWAS were analysed, reporting over 300 loci interacting with environmental factors. Alcohol consumption, sleep time, smoking habit and physical activity were the most studied environmental factors with significant G × E interactions. The implementation of more comprehensive GWAS will provide a better understanding of the metabolic processes that determine individual responses to environmental exposures and their association with the development of chronic diseases such as obesity and the metabolic syndrome. This will facilitate the development of precision approaches for better prevention, management and treatment of these diseases.
Collapse
Affiliation(s)
- Rodrigo San-Cristobal
- grid.23856.3a0000 0004 1936 8390Centre Nutrition, Santé Et Société (NUTRISS), Institut Sur La Nutrition Et Les Aliments Fonctionnels (INAF), Université Laval, Québec, QC Canada ,grid.23856.3a0000 0004 1936 8390School of Nutrition, Université Laval, Quebec, QC G1V 0A6 Canada
| | - Juan de Toro-Martín
- grid.23856.3a0000 0004 1936 8390Centre Nutrition, Santé Et Société (NUTRISS), Institut Sur La Nutrition Et Les Aliments Fonctionnels (INAF), Université Laval, Québec, QC Canada ,grid.23856.3a0000 0004 1936 8390School of Nutrition, Université Laval, Quebec, QC G1V 0A6 Canada
| | - Marie-Claude Vohl
- grid.23856.3a0000 0004 1936 8390Centre Nutrition, Santé Et Société (NUTRISS), Institut Sur La Nutrition Et Les Aliments Fonctionnels (INAF), Université Laval, Québec, QC Canada ,grid.23856.3a0000 0004 1936 8390School of Nutrition, Université Laval, Quebec, QC G1V 0A6 Canada
| |
Collapse
|
21
|
Multi-Omics Interpretation of Anti-Aging Mechanisms for ω-3 Fatty Acids. Genes (Basel) 2021; 12:genes12111691. [PMID: 34828297 PMCID: PMC8625527 DOI: 10.3390/genes12111691] [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: 09/05/2021] [Revised: 10/16/2021] [Accepted: 10/22/2021] [Indexed: 11/16/2022] Open
Abstract
Aging is one of the hottest topics in biomedicine. Previous research suggested that ω-3 fatty acids have preventive effects on aging. However, most of previous studies on the anti-aging effects of ω-3 fatty acids are focused on clinical observations, and the anti-aging mechanisms of ω-3 fatty acids have not been fully elucidated. This stimulated our interest to use multi-omics data related to ω-3 fatty acids in order to interpret the anti-aging mechanisms of ω-3 fatty acids. First, we found that ω-3 fatty acids can affect methylation levels and expression levels of genes associated with age-related diseases or pathways in humans. Then, a Mendelian randomization analysis was conducted to determine whether there is a causal relationship between the effect of ω-3 fatty acids on blood lipid levels and variation in the gut microbiome. Our results indicate that the impact of ω-3 fatty acids on aging is partially mediated by the gut microbiome (including Actinobacteria, Bifidobacteria and Streptococcus). In conclusion, this study provides deeper insights into the anti-aging mechanisms of ω-3 fatty acids and supports the dietary supplementation of ω-3 fatty acids in aging prevention.
Collapse
|
22
|
Hartiala JA, Hilser JR, Biswas S, Lusis AJ, Allayee H. Gene-Environment Interactions for Cardiovascular Disease. Curr Atheroscler Rep 2021; 23:75. [PMID: 34648097 PMCID: PMC8903169 DOI: 10.1007/s11883-021-00974-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE OF REVIEW We provide an overview of recent findings with respect to gene-environment (GxE) interactions for cardiovascular disease (CVD) risk and discuss future opportunities for advancing the field. RECENT FINDINGS Over the last several years, GxE interactions for CVD have mostly been identified for smoking and coronary artery disease (CAD) or related risk factors. By comparison, there is more limited evidence for GxE interactions between CVD outcomes and other exposures, such as physical activity, air pollution, diet, and sex. The establishment of large consortia and population-based cohorts, in combination with new computational tools and mouse genetics platforms, can potentially overcome some of the limitations that have hindered human GxE interaction studies and reveal additional association signals for CVD-related traits. The identification of novel GxE interactions is likely to provide a better understanding of the pathogenesis and genetic liability of CVD, with significant implications for healthy lifestyles and therapeutic strategies.
Collapse
Affiliation(s)
- Jaana A Hartiala
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, CSC202, Los Angeles, CA, 90033, USA
| | - James R Hilser
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, CSC202, Los Angeles, CA, 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Subarna Biswas
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, CSC202, Los Angeles, CA, 90033, USA
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Aldons J Lusis
- Department of Medicine, David Geffen School of Medicine of UCLA, Los Angeles, CA, 90095, USA
- Department of Microbiology, David Geffen School of Medicine of UCLA, Los Angeles, CA, 90095, USA
- Department of Human Genetics, David Geffen School of Medicine of UCLA, Los Angeles, CA, 90095, USA
| | - Hooman Allayee
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, CSC202, Los Angeles, CA, 90033, USA.
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
23
|
Secci R, Hartmann A, Walter M, Grabe HJ, Van der Auwera-Palitschka S, Kowald A, Palmer D, Rimbach G, Fuellen G, Barrantes I. Biomarkers of geroprotection and cardiovascular health: An overview of omics studies and established clinical biomarkers in the context of diet. Crit Rev Food Sci Nutr 2021; 63:2426-2446. [PMID: 34648415 DOI: 10.1080/10408398.2021.1975638] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The slowdown, inhibition, or reversal of age-related decline (as a composite of disease, dysfunction, and, ultimately, death) by diet or natural compounds can be defined as dietary geroprotection. While there is no single reliable biomarker to judge the effects of dietary geroprotection, biomarker signatures based on omics (epigenetics, gene expression, microbiome composition) are promising candidates. Recently, omic biomarkers started to supplement established clinical ones such as lipid profiles and inflammatory cytokines. In this review, we focus on human data. We first summarize the current take on genetic biomarkers based on epidemiological studies. However, most of the remaining biomarkers that we describe, whether omics-based or clinical, are related to intervention studies. Then, because of their promising potential in the context of dietary geroprotection, we focus on the effects of berry-based interventions, which up to now have been mostly described employing clinical markers. We provide an aggregation and tabulation of all the recent systematic reviews and meta-analyses that we could find related to this topic. Finally, we present evidence for the importance of the "nutribiography," that is, the influence that an individual's history of diet and natural compound consumption can have on the effects of dietary geroprotection.
Collapse
Affiliation(s)
- Riccardo Secci
- Junior Research Group Translational Bioinformatics, Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
| | - Alexander Hartmann
- Institute for Clinical Chemistry and Laboratory Medicine, University Medical Center Rostock, University of Rostock, Rostock, Germany
| | - Michael Walter
- Institute for Clinical Chemistry and Laboratory Medicine, University Medical Center Rostock, University of Rostock, Rostock, Germany.,Institute of Laboratory Medicine, Clinical Chemistry, and Pathobiochemistry, Charite University Medical Center, Berlin, Germany
| | - Hans Jörgen Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany.,German Centre for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Greifswald, Germany
| | - Sandra Van der Auwera-Palitschka
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany.,German Centre for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Greifswald, Germany
| | - Axel Kowald
- Institute for Biostatistics and Informatics in Medicine and Aging Research, Rostock University Medical Center, Rostock, Germany
| | - Daniel Palmer
- Institute for Biostatistics and Informatics in Medicine and Aging Research, Rostock University Medical Center, Rostock, Germany
| | - Gerald Rimbach
- Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Georg Fuellen
- Institute for Biostatistics and Informatics in Medicine and Aging Research, Rostock University Medical Center, Rostock, Germany
| | - Israel Barrantes
- Junior Research Group Translational Bioinformatics, Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
24
|
Raggi P. Healthy ageing to guide targeted versus universal treatment: A different approach to prevention. Atherosclerosis 2021; 326:45-46. [PMID: 33906730 DOI: 10.1016/j.atherosclerosis.2021.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 04/14/2021] [Indexed: 11/17/2022]
Affiliation(s)
- Paolo Raggi
- Department of Medicine, University of Alberta, 5A9-014, 11220 83rd Avenue NW, Edmonton, AB, T6G 2B7, Canada.
| |
Collapse
|