1
|
Akbary Moghaddam V, Acharya S, Schwaiger-Haber M, Liao S, Jung WJ, Thyagarajan B, Shriver LP, Daw EW, Saccone NL, An P, Brent MR, Patti GJ, Province MA. Construction of Multi-Modal Transcriptome-Small Molecule Interaction Networks from High-Throughput Measurements to Study Human Complex Traits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.22.634403. [PMID: 39896668 PMCID: PMC11785221 DOI: 10.1101/2025.01.22.634403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Small molecules (SMs) are integral to biological processes, influencing metabolism, homeostasis, and regulatory networks. Despite their importance, a significant knowledge gap exists regarding their downstream effects on biological pathways and gene expression, largely due to differences in scale, variability, and noise between untargeted metabolomics and sequencing-based technologies. To address these challenges, we developed a multi-omics framework comprising a machine learning-based protocol for data processing, a semi-supervised network inference approach, and network-guided analysis of complex traits. The ML protocol harmonized metabolomic, lipidomic, and transcriptomic data through batch correction, principal component analysis, and regression-based adjustments, enabling unbiased and effective integration. Building on this, we proposed a semi-supervised method to construct transcriptome-SM interaction networks (TSI-Nets) by selectively integrating SM profiles into gene-level networks using a meta-analytic approach that accounts for scale differences and missing data across omics layers. Benchmarking against three conventional unsupervised methods demonstrated the superiority of our approach in generating diverse, biologically relevant, and robust networks. While single-omics analyses identified 18 significant genes and 3 significant SMs associated with insulin sensitivity (IS), network-guided analysis revealed novel connections between these markers. The top-ranked module highlighted a cross-talk between fiber-degrading gut microbiota and immune regulatory pathways, inferred by the interaction of the protective SM, N-acetylglycine (NAG), with immune genes (FCER1A, HDC, MS4A2, and CPA3), linked to improved IS and reduced obesity and inflammation. Together, this framework offers a robust and scalable solution for multi-modal network inference and analysis, advancing SM pathway discovery and their implications for human health. Leveraging data from a population of thousands of individuals with extended longevity, the inferred TSI-Nets demonstrate generalizability across diverse conditions and complex traits. These networks are publicly available as a resource for the research community.
Collapse
Affiliation(s)
- Vaha Akbary Moghaddam
- Department of Genetics, School of Medicine, Washington University in St. Louis, MO, USA
| | - Sandeep Acharya
- Division of Computational & Data Sciences, McKelvey School of Engineering, Washington University in St. Louis, MO, USA
| | | | - Shu Liao
- Department of Computer Science & Engineering, McKelvey School of Engineering, Washington University in St. Louis, MO, USA
| | - Wooseok J Jung
- Department of Computer Science & Engineering, McKelvey School of Engineering, Washington University in St. Louis, MO, USA
| | - Bharat Thyagarajan
- Department of Laboratory Medicine & Pathology, School of Medicine, University of Minnesota, MN, USA
| | - Leah P Shriver
- Department of Chemistry, School of Arts & Sciences, Washington University in St. Louis, MO, USA
| | - E Warwick Daw
- Department of Genetics, School of Medicine, Washington University in St. Louis, MO, USA
| | - Nancy L Saccone
- Department of Genetics, School of Medicine, Washington University in St. Louis, MO, USA
| | - Ping An
- Department of Genetics, School of Medicine, Washington University in St. Louis, MO, USA
| | - Michael R Brent
- Department of Genetics, School of Medicine, Washington University in St. Louis, MO, USA
- Department of Computer Science & Engineering, McKelvey School of Engineering, Washington University in St. Louis, MO, USA
| | - Gary J Patti
- Department of Chemistry, School of Arts & Sciences, Washington University in St. Louis, MO, USA
| | - Michael A Province
- Department of Genetics, School of Medicine, Washington University in St. Louis, MO, USA
| |
Collapse
|
2
|
Sarnowski C, Huan T, Ma Y, Joehanes R, Beiser A, DeCarli CS, Heard-Costa NL, Levy D, Lin H, Liu CT, Liu C, Meigs JB, Satizabal CL, Florez JC, Hivert MF, Dupuis J, De Jager PL, Bennett DA, Seshadri S, Morrison AC. Multi-tissue epigenetic analysis identifies distinct associations underlying insulin resistance and Alzheimer's disease at CPT1A locus. Clin Epigenetics 2023; 15:173. [PMID: 37891690 PMCID: PMC10612362 DOI: 10.1186/s13148-023-01589-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/20/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Insulin resistance (IR) is a major risk factor for Alzheimer's disease (AD) dementia. The mechanisms by which IR predisposes to AD are not well-understood. Epigenetic studies may help identify molecular signatures of IR associated with AD, thus improving our understanding of the biological and regulatory mechanisms linking IR and AD. METHODS We conducted an epigenome-wide association study of IR, quantified using the homeostatic model assessment of IR (HOMA-IR) and adjusted for body mass index, in 3,167 participants from the Framingham Heart Study (FHS) without type 2 diabetes at the time of blood draw used for methylation measurement. We identified DNA methylation markers associated with IR at the genome-wide level accounting for multiple testing (P < 1.1 × 10-7) and evaluated their association with neurological traits in participants from the FHS (N = 3040) and the Religious Orders Study/Memory and Aging Project (ROSMAP, N = 707). DNA methylation profiles were measured in blood (FHS) or dorsolateral prefrontal cortex (ROSMAP) using the Illumina HumanMethylation450 BeadChip. Linear regressions (ROSMAP) or mixed-effects models accounting for familial relatedness (FHS) adjusted for age, sex, cohort, self-reported race, batch, and cell type proportions were used to assess associations between DNA methylation and neurological traits accounting for multiple testing. RESULTS We confirmed the strong association of blood DNA methylation with IR at three loci (cg17901584-DHCR24, cg17058475-CPT1A, cg00574958-CPT1A, and cg06500161-ABCG1). In FHS, higher levels of blood DNA methylation at cg00574958 and cg17058475 were both associated with lower IR (P = 2.4 × 10-11 and P = 9.0 × 10-8), larger total brain volumes (P = 0.03 and P = 9.7 × 10-4), and smaller log lateral ventricular volumes (P = 0.07 and P = 0.03). In ROSMAP, higher levels of brain DNA methylation at the same two CPT1A markers were associated with greater risk of cognitive impairment (P = 0.005 and P = 0.02) and higher AD-related indices (CERAD score: P = 5 × 10-4 and 0.001; Braak stage: P = 0.004 and P = 0.01). CONCLUSIONS Our results suggest potentially distinct epigenetic regulatory mechanisms between peripheral blood and dorsolateral prefrontal cortex tissues underlying IR and AD at CPT1A locus.
Collapse
Affiliation(s)
- Chloé Sarnowski
- 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.
| | - Tianxiao Huan
- Population Sciences Branch, National Heart, Lung and Blood Institutes of Health, Bethesda, MD, USA
| | - Yiyi Ma
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Roby Joehanes
- Population Sciences Branch, National Heart, Lung and Blood Institutes of Health, Bethesda, MD, USA
- The Framingham Heart Study, Framingham, MA, USA
| | - Alexa Beiser
- The Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | | | - Nancy L Heard-Costa
- The Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Daniel Levy
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- The Framingham Heart Study, Framingham, MA, USA
| | - Honghuang Lin
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Ching-Ti Liu
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Chunyu Liu
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - James B Meigs
- Division of General Internal Medicine, 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
| | - Claudia L Satizabal
- The Framingham Heart Study, Framingham, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Population Health Sciences, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jose C Florez
- 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
- Center for Genomic Medicine and Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Marie-France Hivert
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Harvard University, Boston, MA, USA
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Josée Dupuis
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
- Department of Epidemiology, Biostatistics and Occupational Health, School of Population and Global Health, McGill University, Montreal, Canada
| | - Philip L De Jager
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Sudha Seshadri
- The Framingham Heart Study, Framingham, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Alanna C Morrison
- 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
| |
Collapse
|
3
|
Lin LC, Liu ZY, Yang JJ, Zhao JY, Tao H. m6A epitranscriptomic modification in diabetic microvascular complications. Trends Pharmacol Sci 2023; 44:S0165-6147(23)00215-8. [PMID: 39492320 DOI: 10.1016/j.tips.2023.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/21/2023] [Accepted: 09/29/2023] [Indexed: 11/05/2024]
Abstract
N6-methyladenosine (m6A) modifications are modulated by m6A methyltransferases, m6A demethylases, and m6A-binding proteins. The dynamic and reversible patterns of m6A modification control cell fate programming by regulating RNA splicing, translation, and decay. Emerging evidence demonstrates that m6A modification of coding and noncoding RNAs exerts crucial effects that influence the pathogenesis of diabetic microvascular complications that include diabetic cardiomyopathy, diabetic nephropathy, diabetic retinopathy, diabetic neuropathy, and diabetic dermatosis. In this review, we summarize the roles of m6A modification and m6A modification-related enzymes in diabetic microvascular complications and discuss potential m6A modification-related enzyme-targeting therapeutic strategies.
Collapse
Affiliation(s)
- Li-Chan Lin
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Zhi-Yan Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Jing-Jing Yang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| | - Jian-Yuan Zhao
- Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Hui Tao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China; Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| |
Collapse
|
4
|
Yao H, Zhou Y, Li T, Li Y, Li F, Zhang G, Fu X, Kang Y, Wu Q. Bioinformatic identification and experiment validation revealed that ACTG1 is a promising prognostic signature and therapeutic target for sepsis. J Leukoc Biol 2023; 114:325-334. [PMID: 37368839 DOI: 10.1093/jleuko/qiad072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 05/24/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
In the intensive care unit, sepsis is a prevalent clinical syndrome (i.e. the final pathway to death from most infections). Peripheral blood gene expression profiling is becoming more and more accepted as a potential diagnostic or prognostic tool. This work aimed to recognize genes related to sepsis, providing potential translational therapeutic targets. RNA sequencing was performed on peripheral blood mononuclear cells from 20 healthy control subjects and 51 sepsis patients. Weighted gene coexpression network analysis was employed to pick out sepsis-related and immunocyte-related gene modules. Genes in the yellow module are primarily involved in excessive inflammation and immune suppression. STRING and Cytoscape were combined to identify ACTG1 and IQGAP1 as hub genes with highest connective degree, and prognostic predication value of ACTG1 was confirmed. Both univariate and multivariate logistic regression analyses were carried out. ACTG1 messenger RNA expression was increased in animal and in cell-related sepsis models. Small interfering RNA revealed decreasing ACTG1 can reduce the in vitro sepsis model apoptosis. We have authenticated ACTG1 as a reliable signature of a poor outcome of sepsis and promising therapeutic targets for sepsis.
Collapse
Affiliation(s)
- Hua Yao
- Department of Critical Care Medicine, West China Hospital, Sichuan University, No.37 Guoxue Alley, Wuhou District, Chengdu, 610041, Sichuan, China
| | - Yue Zhou
- Department of Critical Care Medicine, West China Hospital, Sichuan University, No.37 Guoxue Alley, Wuhou District, Chengdu, 610041, Sichuan, China
| | - Tingting Li
- Department of Critical Care Medicine, West China Hospital, Sichuan University, No.37 Guoxue Alley, Wuhou District, Chengdu, 610041, Sichuan, China
| | - Yao Li
- Department of Critical Care Medicine, West China Hospital, Sichuan University, No.37 Guoxue Alley, Wuhou District, Chengdu, 610041, Sichuan, China
| | - Fan Li
- Department of Critical Care Medicine, West China Hospital, Sichuan University, No.37 Guoxue Alley, Wuhou District, Chengdu, 610041, Sichuan, China
| | - Geng Zhang
- Department of Critical Care Medicine, West China Hospital, Sichuan University, No.37 Guoxue Alley, Wuhou District, Chengdu, 610041, Sichuan, China
| | - Xin Fu
- Department of Critical Care Medicine, West China Hospital, Sichuan University, No.37 Guoxue Alley, Wuhou District, Chengdu, 610041, Sichuan, China
| | - Yan Kang
- Department of Critical Care Medicine, West China Hospital, Sichuan University, No.37 Guoxue Alley, Wuhou District, Chengdu, 610041, Sichuan, China
| | - Qin Wu
- Department of Critical Care Medicine, West China Hospital, Sichuan University, No.37 Guoxue Alley, Wuhou District, Chengdu, 610041, Sichuan, China
| |
Collapse
|
5
|
Stokes T, Cen HH, Kapranov P, Gallagher IJ, Pitsillides AA, Volmar C, Kraus WE, Johnson JD, Phillips SM, Wahlestedt C, Timmons JA. Transcriptomics for Clinical and Experimental Biology Research: Hang on a Seq. ADVANCED GENETICS (HOBOKEN, N.J.) 2023; 4:2200024. [PMID: 37288167 PMCID: PMC10242409 DOI: 10.1002/ggn2.202200024] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Indexed: 06/09/2023]
Abstract
Sequencing the human genome empowers translational medicine, facilitating transcriptome-wide molecular diagnosis, pathway biology, and drug repositioning. Initially, microarrays are used to study the bulk transcriptome; but now short-read RNA sequencing (RNA-seq) predominates. Positioned as a superior technology, that makes the discovery of novel transcripts routine, most RNA-seq analyses are in fact modeled on the known transcriptome. Limitations of the RNA-seq methodology have emerged, while the design of, and the analysis strategies applied to, arrays have matured. An equitable comparison between these technologies is provided, highlighting advantages that modern arrays hold over RNA-seq. Array protocols more accurately quantify constitutively expressed protein coding genes across tissue replicates, and are more reliable for studying lower expressed genes. Arrays reveal long noncoding RNAs (lncRNA) are neither sparsely nor lower expressed than protein coding genes. Heterogeneous coverage of constitutively expressed genes observed with RNA-seq, undermines the validity and reproducibility of pathway analyses. The factors driving these observations, many of which are relevant to long-read or single-cell sequencing are discussed. As proposed herein, a reappreciation of bulk transcriptomic methods is required, including wider use of the modern high-density array data-to urgently revise existing anatomical RNA reference atlases and assist with more accurate study of lncRNAs.
Collapse
Affiliation(s)
- Tanner Stokes
- Faculty of ScienceMcMaster UniversityHamiltonL8S 4L8Canada
| | - Haoning Howard Cen
- Life Sciences InstituteUniversity of British ColumbiaVancouverV6T 1Z3Canada
| | | | - Iain J Gallagher
- School of Applied SciencesEdinburgh Napier UniversityEdinburghEH11 4BNUK
| | | | | | | | - James D. Johnson
- Life Sciences InstituteUniversity of British ColumbiaVancouverV6T 1Z3Canada
| | | | | | - James A. Timmons
- Miller School of MedicineUniversity of MiamiMiamiFL33136USA
- William Harvey Research InstituteQueen Mary University LondonLondonEC1M 6BQUK
- Augur Precision Medicine LTDStirlingFK9 5NFUK
| |
Collapse
|
6
|
Wu X, Wang W, Fan S, You L, Li F, Zhang X, Wu H, Tang J, Qi Y, Feng W, Yan L, Ren M. U-shaped association between serum IGF2BP3 and T2DM: A cross-sectional study in Chinese population. J Diabetes 2023; 15:349-361. [PMID: 36891946 PMCID: PMC10101838 DOI: 10.1111/1753-0407.13378] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 02/03/2023] [Accepted: 02/20/2023] [Indexed: 03/10/2023] Open
Abstract
OBJECTIVE To clarify the expression of N6-methyladenosine (m6 A) modulators involved in the pathogenesis of type 2 diabetes mellitus (T2DM). We further explored the association of serum insulin-like growth factor 2 mRNA-binding proteins 3 (IGF2BP3) levels and odds of T2DM in a high-risk population. METHODS The gene expression data set GSE25724 was obtained from the Gene Expression Omnibus, and a cluster heatmap was generated by using the R package ComplexHeatmap. Differential expression analysis for 13 m6 A RNA methylation regulators between nondiabetic controls and T2DM subjects was performed using an unpaired t test. A cross-sectional design, including 393 subjects (131 patients with newly diagnosed T2DM, 131 age- and sex-matched subjects with prediabetes, and 131 healthy controls), was carried out. The associations between serum IGF2BP3 concentrations and T2DM were modeled by restricted cubic spline and logistic regression models. RESULTS Two upregulated (IGF2BP2 and IGF2BP3) and 5 downregulated (methyltransferase-like 3 [METTL3], alkylation repair homolog protein 1 [ALKBH1], YTH domain family 2 [YTHDF2], YTHDF3, and heterogeneous nuclear ribonucleoprotein [HNRNPC]) m6 A-related genes were found in islet samples of T2DM patients. A U-shaped association existed between serum IGF2BP3 levels and odds of T2DM according to cubic natural spline analysis models, after adjustment for body mass index, waist circumference, diastolic blood pressure, total cholesterol, and triglyeride. Multivariate logistic regression showed that progressively higher odds of T2DM were observed when serum IGF2BP3 levels were below 0.62 ng/mL (odds ratio 3.03 [95% confidence interval 1.23-7.47]) in model 4. CONCLUSION Seven significantly altered m6 A RNA methylation genes were identified in T2DM. There was a U-shaped association between serum IGF2BP3 levels and odds of T2DM in the general Chinese adult population. This study provides important evidence for further examination of the role of m6 A RNA methylation, especially serum IGF2BP3 in T2DM risk assessment.
Collapse
Affiliation(s)
- Xiaoying Wu
- Department of EndocrinologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouPeople's Republic of China
- Department of EndocrinologyNational Center of Gerontology, Beijing Hospital, Peking University Fifth School of Clinical MedicineBeijingChina
| | - Wei Wang
- Department of EndocrinologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouPeople's Republic of China
| | - Shujin Fan
- Department of EndocrinologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouPeople's Republic of China
| | - Lili You
- Department of EndocrinologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouPeople's Republic of China
| | - Feng Li
- Department of EndocrinologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouPeople's Republic of China
| | - Xiaoyun Zhang
- Department of EndocrinologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouPeople's Republic of China
| | - Hongshi Wu
- Department of EndocrinologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouPeople's Republic of China
| | - Juying Tang
- Department of EndocrinologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouPeople's Republic of China
| | - Yiqin Qi
- Department of EndocrinologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouPeople's Republic of China
| | - Wanting Feng
- Department of EndocrinologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouPeople's Republic of China
| | - Li Yan
- Department of EndocrinologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouPeople's Republic of China
| | - Meng Ren
- Department of EndocrinologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouPeople's Republic of China
| |
Collapse
|
7
|
Harary D, Akinyemi A, Charron MJ, Fuloria M. Fetal Growth and Intrauterine Epigenetic Programming of Obesity and Cardiometabolic Disease. Neoreviews 2022; 23:e363-e372. [PMID: 35641462 PMCID: PMC10100845 DOI: 10.1542/neo.23-6-e363] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Epidemiologic studies have shown an association between an adverse intrauterine environment (eg, exposure to malnutrition) and an increased risk of developing cardiometabolic disease in adulthood. These studies laid the foundation for the developmental origins of health and disease hypothesis, which states that limited nutrient supply to the fetus results in physiologic and metabolic adaptations that favor survival but result in unfavorable consequences in the offspring if there is excess nutrition after birth. This discrepancy in the pre- and postnatal milieus, perceived as stress by the offspring, may confer an increased risk of developing cardiometabolic disease later in life. Thus, early life exposures result in programming or changes in cellular memory that have effects on health throughout the life course. One of the mechanisms by which programming occurs is via epigenetic modifications of genes, processes that result in functionally relevant changes in genes (ie, gene expression) without an alteration in the genotype. In this review, we will describe how fetal exposures, including under- and overnutrition, affect neonatal and childhood growth and the future risk for cardiometabolic disease.
Collapse
Affiliation(s)
- David Harary
- Department of Pediatrics, Division of Neonatology, Children's Hospital at Montefiore, Bronx, NY
| | | | - Maureen J Charron
- Departments of †Biochemistry
- Obstetrics & Gynecology and Women's Health, and
- Medicine, Division of Endocrinology, Albert Einstein College of Medicine, Bronx, NY
| | - Mamta Fuloria
- Department of Pediatrics, Division of Neonatology, Children's Hospital at Montefiore, Bronx, NY
| |
Collapse
|
8
|
Zeng F, Zhao C, Li N, Gao X, Pan YY, Liu B, Pang J. Effects of Alkaloid-Rich Extracts Obtained from Grifola frondosa on Gut Microbiota and Glucose Homeostasis in Rats. Food Funct 2022; 13:2729-2742. [DOI: 10.1039/d1fo04062f] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Grifola frondosa (GF), also known as maitake (a type of mushroom), has been widely used as a food item and it exhibits various health-beneficial hypoglycemic activities. Rats fed with a...
Collapse
|
9
|
Nayor M, Shah SH, Murthy V, Shah RV. Molecular Aspects of Lifestyle and Environmental Effects in Patients With Diabetes: JACC Focus Seminar. J Am Coll Cardiol 2021; 78:481-495. [PMID: 34325838 DOI: 10.1016/j.jacc.2021.02.070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/07/2021] [Accepted: 02/01/2021] [Indexed: 01/04/2023]
Abstract
Diabetes is characterized as an integrated condition of dysregulated metabolism across multiple tissues, with well-established consequences on the cardiovascular system. Recent advances in precision phenotyping in biofluids and tissues in large human observational and interventional studies have afforded a unique opportunity to translate seminal findings in models and cellular systems to patients at risk for diabetes and its complications. Specifically, techniques to assay metabolites, proteins, and transcripts, alongside more recent assessment of the gut microbiome, underscore the complexity of diabetes in patients, suggesting avenues for precision phenotyping of risk, response to intervention, and potentially novel therapies. In addition, the influence of external factors and inputs (eg, activity, diet, medical therapies) on each domain of molecular characterization has gained prominence toward better understanding their role in prevention. Here, the authors provide a broad overview of the role of several of these molecular domains in human translational investigation in diabetes.
Collapse
Affiliation(s)
- Matthew Nayor
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA. https://twitter.com/MattNayor
| | - Svati H Shah
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA; Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA. https://twitter.com/SvatiShah
| | - Venkatesh Murthy
- Division of Cardiovascular Medicine, Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA; Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan, USA. https://twitter.com/venkmurthy
| | - Ravi V Shah
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
10
|
The biological function of IGF2BPs and their role in tumorigenesis. Invest New Drugs 2021; 39:1682-1693. [PMID: 34251559 DOI: 10.1007/s10637-021-01148-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/30/2021] [Indexed: 01/09/2023]
Abstract
The insulin-like growth factor-2 mRNA-binding proteins (IGF2BPs) pertain to a highly conservative RNA-binding family that works as a post-transcriptional fine-tuner for target transcripts. Emerging evidence suggests that IGF2BPs regulate RNA processing and metabolism, including stability, translation, and localization, and are involved in various cellular functions and pathophysiologies. In this review, we summarize the roles and molecular mechanisms of IGF2BPs in cancer development and progression. We mainly discuss the functional relevance of IGF2BPs in embryo development, neurogenesis, metabolism, RNA processing, and tumorigenesis. Understanding IGF2BPs role in tumor progression will provide new insight into cancer pathophysiology.
Collapse
|
11
|
Nonnecke EB, Castillo PA, Dugan AE, Almalki F, Underwood MA, De La Motte CA, Yuan W, Lu W, Shen B, Johansson MEV, Kiessling LL, Hollox EJ, Lönnerdal B, Bevins CL. Human intelectin-1 (ITLN1) genetic variation and intestinal expression. Sci Rep 2021; 11:12889. [PMID: 34145348 PMCID: PMC8213764 DOI: 10.1038/s41598-021-92198-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/07/2021] [Indexed: 12/13/2022] Open
Abstract
Intelectins are ancient carbohydrate binding proteins, spanning chordate evolution and implicated in multiple human diseases. Previous GWAS have linked SNPs in ITLN1 (also known as omentin) with susceptibility to Crohn's disease (CD); however, analysis of possible functional significance of SNPs at this locus is lacking. Using the Ensembl database, pairwise linkage disequilibrium (LD) analyses indicated that several disease-associated SNPs at the ITLN1 locus, including SNPs in CD244 and Ly9, were in LD. The alleles comprising the risk haplotype are the major alleles in European (67%), but minor alleles in African superpopulations. Neither ITLN1 mRNA nor protein abundance in intestinal tissue, which we confirm as goblet-cell derived, was altered in the CD samples overall nor when samples were analyzed according to genotype. Moreover, the missense variant V109D does not influence ITLN1 glycan binding to the glycan β-D-galactofuranose or protein-protein oligomerization. Taken together, our data are an important step in defining the role(s) of the CD-risk haplotype by determining that risk is unlikely to be due to changes in ITLN1 carbohydrate recognition, protein oligomerization, or expression levels in intestinal mucosa. Our findings suggest that the relationship between the genomic data and disease arises from changes in CD244 or Ly9 biology, differences in ITLN1 expression in other tissues, or an alteration in ITLN1 interaction with other proteins.
Collapse
Affiliation(s)
- Eric B Nonnecke
- Department of Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA, 95616, USA.
| | - Patricia A Castillo
- Department of Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA, 95616, USA
- Elanco Animal Health, Fort Dodge, IA, 50501, USA
| | - Amanda E Dugan
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Faisal Almalki
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
- Medical Laboratories Technology Department, College of Applied Medical Sciences, Taibah University, Almadinah Almunwarah, Saudi Arabia
| | - Mark A Underwood
- Department of Pediatrics, School of Medicine, University of California, Davis, Sacramento, CA, 95817, USA
| | - Carol A De La Motte
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, 44195, USA
| | - Weirong Yuan
- Institute of Human Virology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Wuyuan Lu
- Institute of Human Virology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Fudan University, Shanghai, China
| | - Bo Shen
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
- Department of Surgery, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Malin E V Johansson
- Institute of Biomedicine, Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden
| | - Laura L Kiessling
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Edward J Hollox
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Bo Lönnerdal
- Department of Nutrition, University of California, Davis, Davis, CA, 95616, USA
| | - Charles L Bevins
- Department of Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA, 95616, USA.
| |
Collapse
|
12
|
Regué L, Zhao L, Ji F, Wang H, Avruch J, Dai N. RNA m6A reader IMP2/IGF2BP2 promotes pancreatic β-cell proliferation and insulin secretion by enhancing PDX1 expression. Mol Metab 2021; 48:101209. [PMID: 33705986 PMCID: PMC8076713 DOI: 10.1016/j.molmet.2021.101209] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 02/23/2021] [Accepted: 03/05/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) is a common metabolic disease. Variants in human IGF2 mRNA binding protein 2 (IMP2/IGF2BP2) are associated with increased risk of T2D. IMP2 contributes to T2D susceptibility primarily through effects on insulin secretion. However, the underlying mechanism is not known. METHODS To understand the role of IMP2 in insulin secretion and T2D pathophysiology, we generated Imp2 pancreatic β-cell specific knockout mice (βIMP2KO) by recombining the Imp2flox allele with Cre recombinase driven by the rat insulin 2 promoter. We further characterized metabolic phenotypes of βIMP2KO mice and assessed their β-cell functions. RESULTS The deletion of IMP2 in pancreatic β-cells leads to reduced compensatory β-cell proliferation and function. Mechanically, IMP2 directly binds to Pdx1 mRNA and stimulates its translation in an m6A dependent manner. Moreover, IMP2 orchestrates IGF2-AKT-GSK3β-PDX1 signaling to stable PDX1 polypeptides. In human EndoC-βH1 cells, the over-expression of IMP2 is capable to enhance cell proliferation, PDX1 protein level and insulin secretion. CONCLUSION Our work therefore reveals IMP2 as a critical regulator of pancreatic β-cell proliferation and function; highlights the importance of posttranscriptional gene expression in T2D pathology.
Collapse
MESH Headings
- Adenosine/analogs & derivatives
- Adenosine/metabolism
- Animals
- Cell Line
- Cell Proliferation/genetics
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diet, High-Fat/adverse effects
- Disease Models, Animal
- Gene Knockout Techniques
- Homeodomain Proteins/metabolism
- Humans
- Insulin Secretion/genetics
- Insulin, Regular, Human/administration & dosage
- Insulin, Regular, Human/genetics
- Insulin, Regular, Human/metabolism
- Insulin-Secreting Cells/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Middle Aged
- Promoter Regions, Genetic
- RNA, Messenger/metabolism
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/metabolism
- Rats
- Signal Transduction/genetics
- Trans-Activators/metabolism
- Transfection
Collapse
Affiliation(s)
- Laura Regué
- Department of Molecular Biology and Diabetes Unit of the Medical Services, Massachusetts General Hospital, Boston, 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Liping Zhao
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Fei Ji
- Department of Molecular Biology and Diabetes Unit of the Medical Services, Massachusetts General Hospital, Boston, 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Hua Wang
- The Lundquist Institute, Harbor-UCLA, Torrance, CA, 90502, USA
| | - Joseph Avruch
- Department of Molecular Biology and Diabetes Unit of the Medical Services, Massachusetts General Hospital, Boston, 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Ning Dai
- Department of Molecular Biology and Diabetes Unit of the Medical Services, Massachusetts General Hospital, Boston, 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
13
|
Gui W, Liang J, Lin X, Shi N, Zhu Y, Tan B, Li H. Association of Genetic Variants in IGF2-Related Genes With Risk of Metabolic Syndrome in the Chinese Han Population. Front Endocrinol (Lausanne) 2021; 12:654747. [PMID: 34093434 PMCID: PMC8173176 DOI: 10.3389/fendo.2021.654747] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 04/15/2021] [Indexed: 11/13/2022] Open
Abstract
AIMS To explore associations between polymorphisms of IGF2-related genes including H19, IGF2, IGF2BP2 and IGF2R and Metabolic syndrome (MetS) susceptibility in the Chinese Han population. METHODS 66 subjects with MetS and 257 control subjects were collected for inclusion in a case-control study. PCR-RFLP was used to investigate polymorphisms in the H19, IGF2, IGF2BP2 and IGF2R genes. Elisa was used to detect the serum IGF2 concentrations. RESULTS Females carrying the GG and AG genotypes of rs680 (IGF2) exhibited a lower risk of MetS, compared with those harboring AA (adjusted OR = 0.388, p = 0.027), while GG and AG genotypes were associated with lower fasting glucose and HbA1c. In males, the Waist-to-Hip Ratio (WHR) and the level of TG were significantly higher in GG and AG genotypes than in the AA genotype of rs680 in IGF2. Levels of HDL-c were lower in men with GG and AG genotypes compared with those carrying the AA genotype. Serum IGF2 concentrations did not change among different genotypes. Finally, multifactor dimensionality reduction (MDR) analysis identified interactions between four polymorphisms: rs3741279 (H19), rs680 (IGF2), rs1470579 (IGF2BP2) and rs629849 (IGF2R). CONCLUSIONS Our study suggests that IGF2-related genes including H19, IGF2, IGF2BP2 and IGF2R genes may play pivotal roles in the development of MetS.
Collapse
|
14
|
Deshmukh HA, Madsen AL, Viñuela A, Have CT, Grarup N, Tura A, Mahajan A, Heggie AJ, Koivula RW, De Masi F, Tsirigos KK, Linneberg A, Drivsholm T, Pedersen O, Sørensen TIA, Astrup A, Gjesing AAP, Pavo I, Wood AR, Ruetten H, Jones AG, Koopman ADM, Cederberg H, Rutters F, Ridderstrale M, Laakso M, McCarthy MI, Frayling TM, Ferrannini E, Franks PW, Pearson ER, Mari A, Hansen T, Walker M. Genome-Wide Association Analysis of Pancreatic Beta-Cell Glucose Sensitivity. J Clin Endocrinol Metab 2021; 106:80-90. [PMID: 32944759 PMCID: PMC7765651 DOI: 10.1210/clinem/dgaa653] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 09/14/2020] [Indexed: 11/19/2022]
Abstract
CONTEXT Pancreatic beta-cell glucose sensitivity is the slope of the plasma glucose-insulin secretion relationship and is a key predictor of deteriorating glucose tolerance and development of type 2 diabetes. However, there are no large-scale studies looking at the genetic determinants of beta-cell glucose sensitivity. OBJECTIVE To understand the genetic determinants of pancreatic beta-cell glucose sensitivity using genome-wide meta-analysis and candidate gene studies. DESIGN We performed a genome-wide meta-analysis for beta-cell glucose sensitivity in subjects with type 2 diabetes and nondiabetic subjects from 6 independent cohorts (n = 5706). Beta-cell glucose sensitivity was calculated from mixed meal and oral glucose tolerance tests, and its associations between known glycemia-related single nucleotide polymorphisms (SNPs) and genome-wide association study (GWAS) SNPs were estimated using linear regression models. RESULTS Beta-cell glucose sensitivity was moderately heritable (h2 ranged from 34% to 55%) using SNP and family-based analyses. GWAS meta-analysis identified multiple correlated SNPs in the CDKAL1 gene and GIPR-QPCTL gene loci that reached genome-wide significance, with SNP rs2238691 in GIPR-QPCTL (P value = 2.64 × 10-9) and rs9368219 in the CDKAL1 (P value = 3.15 × 10-9) showing the strongest association with beta-cell glucose sensitivity. These loci surpassed genome-wide significance when the GWAS meta-analysis was repeated after exclusion of the diabetic subjects. After correction for multiple testing, glycemia-associated SNPs in or near the HHEX and IGF2B2 loci were also associated with beta-cell glucose sensitivity. CONCLUSION We show that, variation at the GIPR-QPCTL and CDKAL1 loci are key determinants of pancreatic beta-cell glucose sensitivity.
Collapse
Affiliation(s)
- Harshal A Deshmukh
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Anne Lundager Madsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ana Viñuela
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva
| | - Christian Theil Have
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Andrea Tura
- Institute of Neuroscience, National Research Council, Corso Stati Uniti 4, Padua, Italy
| | - Anubha Mahajan
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Alison J Heggie
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Robert W Koivula
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Skåne University Hospital Malmö, Lund University, 205 02 Malmö, Sweden
| | - Federico De Masi
- Integrative Systems Biology Group, Department of Health Technology, Technical University of Denmark (DTU), Kemitorvet, Building 208, 2800 Kgs. Lyngby, Denmark
| | - Konstantinos K Tsirigos
- Integrative Systems Biology Group, Department of Health Technology, Technical University of Denmark (DTU), Kemitorvet, Building 208, 2800 Kgs. Lyngby, Denmark
| | - Allan Linneberg
- Center for Clinical Research and Disease Prevention, Bispebjerg and Frederiksberg Hospital, The Capital Region, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Drivsholm
- Center for Clinical Research and Disease Prevention, Bispebjerg and Frederiksberg Hospital, The Capital Region, Copenhagen, Denmark
- Section of General Practice, Institute of Public Health, Faculty of Health Sciences, University of Copenhagen, Øster Farimagsgade 5, Copenhagen, Denmark
| | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thorkild I A Sørensen
- Novo Nordisk Foundation Centre for Basic Metabolic Research (Section of Metabolic Genetics), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Public Health (Section of Epidemiology), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Arne Astrup
- Department of Nutrition, Exercise and Sports (NEXS), Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Anette A P Gjesing
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Imre Pavo
- Eli Lilly Regional Operations Ges.m.b.H., Koelblgasse 8–10, Vienna, Austria
| | - Andrew R Wood
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Hartmut Ruetten
- Diabetes Division, Sanofi-Aventis Deutschland GmbH, Frankfurt, 65926 Frankfurt am Main, Germany
| | - Angus G Jones
- NIHR Exeter Clinical Research Facility, University of Exeter Medical School, Exeter, UK
| | - Anitra D M Koopman
- Department of Epidemiology and Biostatistics, VUMC, de Boelelaan 1089a, HV, Amsterdam, the Netherlands
| | - Henna Cederberg
- Department of Endocrinology, Abdominal Centre, Helsinki University Hospital, Helsinki, Finland
| | - Femke Rutters
- Department of Epidemiology and Biostatistics, VUMC, de Boelelaan 1089a, HV, Amsterdam, the Netherlands
| | - Martin Ridderstrale
- Department of Clinical Sciences, Diabetes & Endocrinology Unit, Lund University, Skåne University Hospital Malmö, CRC, 91-12, 205 02, Malmö, Sweden
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Mark I McCarthy
- Oxford Centre for Diabetes Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Tim M Frayling
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter, UK
| | | | - Paul W Franks
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Skåne University Hospital Malmö, Lund University, 205 02 Malmö, Sweden
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, Massachusetts
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Ewan R Pearson
- Division of Population Health & Genomics, School of Medicine, University of Dundee, Dundee, UK
| | - Andrea Mari
- Institute of Neuroscience, National Research Council, Corso Stati Uniti 4, Padua, Italy
- Correspondence and Reprint Requests: Prof Mark Walker, Translational and Clinical Research Institute (Diabetes), The Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH. E-mail: ; Prof Torben Hansen, Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Maersk Tower, Blegdamsvej 3B, 07-8-26, DK-2200, Copenhagen N, Denmark. E-mail: ; Dr Andrea Mari, Institute of Neuroscience, National Research Council, Corso Stati Uniti 4, 35127 Padova, Italy. E-mail:
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Correspondence and Reprint Requests: Prof Mark Walker, Translational and Clinical Research Institute (Diabetes), The Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH. E-mail: ; Prof Torben Hansen, Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Maersk Tower, Blegdamsvej 3B, 07-8-26, DK-2200, Copenhagen N, Denmark. E-mail: ; Dr Andrea Mari, Institute of Neuroscience, National Research Council, Corso Stati Uniti 4, 35127 Padova, Italy. E-mail:
| | - Mark Walker
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Correspondence and Reprint Requests: Prof Mark Walker, Translational and Clinical Research Institute (Diabetes), The Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH. E-mail: ; Prof Torben Hansen, Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Maersk Tower, Blegdamsvej 3B, 07-8-26, DK-2200, Copenhagen N, Denmark. E-mail: ; Dr Andrea Mari, Institute of Neuroscience, National Research Council, Corso Stati Uniti 4, 35127 Padova, Italy. E-mail:
| |
Collapse
|
15
|
Vehmeijer FOL, Küpers LK, Sharp GC, Salas LA, Lent S, Jima DD, Tindula G, Reese S, Qi C, Gruzieva O, Page C, Rezwan FI, Melton PE, Nohr E, Escaramís G, Rzehak P, Heiskala A, Gong T, Tuominen ST, Gao L, Ross JP, Starling AP, Holloway JW, Yousefi P, Aasvang GM, Beilin LJ, Bergström A, Binder E, Chatzi L, Corpeleijn E, Czamara D, Eskenazi B, Ewart S, Ferre N, Grote V, Gruszfeld D, Håberg SE, Hoyo C, Huen K, Karlsson R, Kull I, Langhendries JP, Lepeule J, Magnus MC, Maguire RL, Molloy PL, Monnereau C, Mori TA, Oken E, Räikkönen K, Rifas-Shiman S, Ruiz-Arenas C, Sebert S, Ullemar V, Verduci E, Vonk JM, Xu CJ, Yang IV, Zhang H, Zhang W, Karmaus W, Dabelea D, Muhlhausler BS, Breton CV, Lahti J, Almqvist C, Jarvelin MR, Koletzko B, Vrijheid M, Sørensen TIA, Huang RC, Arshad SH, Nystad W, Melén E, Koppelman GH, London SJ, Holland N, Bustamante M, Murphy SK, Hivert MF, Baccarelli A, Relton CL, Snieder H, Jaddoe VWV, Felix JF. DNA methylation and body mass index from birth to adolescence: meta-analyses of epigenome-wide association studies. Genome Med 2020; 12:105. [PMID: 33239103 PMCID: PMC7687793 DOI: 10.1186/s13073-020-00810-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 11/12/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND DNA methylation has been shown to be associated with adiposity in adulthood. However, whether similar DNA methylation patterns are associated with childhood and adolescent body mass index (BMI) is largely unknown. More insight into this relationship at younger ages may have implications for future prevention of obesity and its related traits. METHODS We examined whether DNA methylation in cord blood and whole blood in childhood and adolescence was associated with BMI in the age range from 2 to 18 years using both cross-sectional and longitudinal models. We performed meta-analyses of epigenome-wide association studies including up to 4133 children from 23 studies. We examined the overlap of findings reported in previous studies in children and adults with those in our analyses and calculated enrichment. RESULTS DNA methylation at three CpGs (cg05937453, cg25212453, and cg10040131), each in a different age range, was associated with BMI at Bonferroni significance, P < 1.06 × 10-7, with a 0.96 standard deviation score (SDS) (standard error (SE) 0.17), 0.32 SDS (SE 0.06), and 0.32 BMI SDS (SE 0.06) higher BMI per 10% increase in methylation, respectively. DNA methylation at nine additional CpGs in the cross-sectional childhood model was associated with BMI at false discovery rate significance. The strength of the associations of DNA methylation at the 187 CpGs previously identified to be associated with adult BMI, increased with advancing age across childhood and adolescence in our analyses. In addition, correlation coefficients between effect estimates for those CpGs in adults and in children and adolescents also increased. Among the top findings for each age range, we observed increasing enrichment for the CpGs that were previously identified in adults (birth Penrichment = 1; childhood Penrichment = 2.00 × 10-4; adolescence Penrichment = 2.10 × 10-7). CONCLUSIONS There were only minimal associations of DNA methylation with childhood and adolescent BMI. With the advancing age of the participants across childhood and adolescence, we observed increasing overlap with altered DNA methylation loci reported in association with adult BMI. These findings may be compatible with the hypothesis that DNA methylation differences are mostly a consequence rather than a cause of obesity.
Collapse
Affiliation(s)
- Florianne O L Vehmeijer
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Room Na-2918, Erasmus MC, PO Box 2040, 3000 CA, Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Leanne K Küpers
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, Groningen, the Netherlands
| | - Gemma C Sharp
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Lucas A Salas
- Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
- ISGlobal, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Samantha Lent
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Dereje D Jima
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA
| | - Gwen Tindula
- Children's Environmental Health Laboratory, Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA, USA
| | - Sarah Reese
- Department of Health and Human Services, Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Cancan Qi
- University of Groningen, University Medical Center Groningen, Department of Pediatric Pulmonology and Pediatric Allergy, Beatrix Children's Hospital, Groningen, The Netherlands
- University Medical Center Groningen GRIAC Research Institute, University of Groningen, Groningen, the Netherlands
| | - Olena Gruzieva
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Centre for Occupational and Environmental Medicine, Region Stockholm, Stockholm, Sweden
| | - Christian Page
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
- Oslo Centre for Biostatistics and Epidemiology, Oslo University Hospital, Oslo, Norway
| | - Faisal I Rezwan
- School of Water, Energy and Environment, Cranfield University, Cranfield, Bedfordshire, UK
- Human Development and Health, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, UK
| | - Philip E Melton
- School of Pharmacy and Biomedical Sciences, Curtin University, Bentley, Western Australia, Australia
- School of Biomedical Sciences, The University of Western Australia, Crawley, Western Austalia, Australia
| | - Ellen Nohr
- Centre for Women's, Family and Child Health, University of South-Eastern Norway, Kongsberg, Norway
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Geòrgia Escaramís
- CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
- Research group on Statistics, Econometrics and Health (GRECS), University of Girona, Girona, Spain
| | - Peter Rzehak
- Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, Ludwig-Maximilians Universität München (LMU), Munich, Germany
| | - Anni Heiskala
- Center for Life Course Health Research, University of Oulu, Oulu, Finland
| | - Tong Gong
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Samuli T Tuominen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Lu Gao
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jason P Ross
- CSIRO Health and Biosecurity, North Ryde, New South Wales, Australia
| | - Anne P Starling
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - John W Holloway
- Human Development and Health, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, UK
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Paul Yousefi
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Gunn Marit Aasvang
- Department of Air Pollution and Noise, Norwegian Institute of Public Health, Oslo, Norway
| | | | - Anna Bergström
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Centre for Occupational and Environmental Medicine, Region Stockholm, Stockholm, Sweden
| | - Elisabeth Binder
- Department of Translational Research in Psychiatry, Max-Planck-Institute of Psychiatry, Munich, Germany
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Leda Chatzi
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Eva Corpeleijn
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, Groningen, the Netherlands
| | - Darina Czamara
- Department of Translational Research in Psychiatry, Max-Planck-Institute of Psychiatry, Munich, Germany
| | - Brenda Eskenazi
- Center for Environmental Research and Children's Health, School of Public Health, University of California, Berkeley, CA, USA
| | - Susan Ewart
- College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| | - Natalia Ferre
- Pediatrics, Nutrition and Development Research Unit, Universitat Rovira i Virgili, IISPV, Reus, Spain
| | - Veit Grote
- Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, Ludwig-Maximilians Universität München (LMU), Munich, Germany
| | - Dariusz Gruszfeld
- Neonatal Department, Children's Memorial Health Institute, Warsaw, Poland
| | - Siri E Håberg
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Cathrine Hoyo
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Karen Huen
- Children's Environmental Health Laboratory, Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA, USA
| | - Robert Karlsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Inger Kull
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
- Sachs' Children and Youth Hospital, Södersjukhuset, Stockholm, Sweden
| | | | - Johanna Lepeule
- Université Grenoble Alpes, Inserm, CNRS, Team of Environmental Epidemiology Applied to Reproduction and Respiratory Health, IAB, Grenoble, France
| | - Maria C Magnus
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Rachel L Maguire
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
- Department of Obstetrics and Gynecology, Duke University Medical Center, Raleigh, NC, USA
| | - Peter L Molloy
- CSIRO Health and Biosecurity, North Ryde, New South Wales, Australia
| | - Claire Monnereau
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Room Na-2918, Erasmus MC, PO Box 2040, 3000 CA, Rotterdam, the Netherlands
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Trevor A Mori
- Medical School, University of Western Australia, Perth, Australia
| | - Emily Oken
- Department of Population Medicine, Harvard Medical School, Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Katri Räikkönen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sheryl Rifas-Shiman
- Department of Population Medicine, Harvard Medical School, Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Carlos Ruiz-Arenas
- ISGlobal, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Sylvain Sebert
- Center for Life Course Health Research, University of Oulu, Oulu, Finland
| | - Vilhelmina Ullemar
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Elvira Verduci
- Department of Pediatrics, San Paolo Hospital, University of Milan, Milan, Italy
| | - Judith M Vonk
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, Groningen, the Netherlands
- University Medical Center Groningen GRIAC Research Institute, University of Groningen, Groningen, the Netherlands
| | - Cheng-Jian Xu
- University of Groningen, University Medical Center Groningen, Department of Pediatric Pulmonology and Pediatric Allergy, Beatrix Children's Hospital, Groningen, The Netherlands
- University Medical Center Groningen GRIAC Research Institute, University of Groningen, Groningen, the Netherlands
- Department of Gastroenterology, Hepatology and Endocrinology, CiiM, Centre for Individualised Infection Medicine, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
| | - Ivana V Yang
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO, USA
| | - Hongmei Zhang
- Division of Epidemiology, Biostatistics, and Environmental Health, University of Memphis, Memphis, TN, USA
| | - Weiming Zhang
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO, USA
| | - Wilfried Karmaus
- Division of Epidemiology, Biostatistics, and Environmental Health, University of Memphis, Memphis, TN, USA
| | - Dana Dabelea
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Carrie V Breton
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jari Lahti
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Turku Institute for Advanced Studies, University of Turku, Turku, Finland
| | - Catarina Almqvist
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Pediatric Allergy and Pulmonology Unit at Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Marjo-Riitta Jarvelin
- Center for Life Course Health Research, University of Oulu, Oulu, Finland
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Unit of Primary Health Care, Oulu University Hospital, OYS, Oulu, Finland
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, London, UK
| | - Berthold Koletzko
- Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, Ludwig-Maximilians Universität München (LMU), Munich, Germany
| | - Martine Vrijheid
- ISGlobal, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Thorkild I A Sørensen
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Department of Public Health, Section of Epidemiology, and The Novo Nordisk Foundation Center for Basic Metabolic Research, Section on Metabolic Genetics, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rae-Chi Huang
- Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - Syed Hasan Arshad
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- David Hide Asthma and Allergy Research Centre, Isle of Wight, UK
| | - Wenche Nystad
- Department of Chronic Diseases and Ageing, Norwegian Institute of Public Health, Oslo, Norway
| | - Erik Melén
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
- Sachs' Children and Youth Hospital, Södersjukhuset, Stockholm, Sweden
| | - Gerard H Koppelman
- University of Groningen, University Medical Center Groningen, Department of Pediatric Pulmonology and Pediatric Allergy, Beatrix Children's Hospital, Groningen, The Netherlands
- University Medical Center Groningen GRIAC Research Institute, University of Groningen, Groningen, the Netherlands
| | - Stephanie J London
- Department of Health and Human Services, Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Nina Holland
- Children's Environmental Health Laboratory, Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA, USA
| | - Mariona Bustamante
- ISGlobal, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Susan K Murphy
- Department of Obstetrics and Gynecology, Duke University Medical Center, Raleigh, NC, USA
| | - Marie-France Hivert
- Department of Population Medicine, Harvard Medical School, Harvard Pilgrim Health Care Institute, Boston, MA, USA
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Universite de Sherbrooke, Sherbrooke, QC, Canada
| | - Andrea Baccarelli
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Caroline L Relton
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Harold Snieder
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, Groningen, the Netherlands
| | - Vincent W V Jaddoe
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Room Na-2918, Erasmus MC, PO Box 2040, 3000 CA, Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Janine F Felix
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Room Na-2918, Erasmus MC, PO Box 2040, 3000 CA, Rotterdam, the Netherlands.
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| |
Collapse
|
16
|
Smoking-related changes in DNA methylation and gene expression are associated with cardio-metabolic traits. Clin Epigenetics 2020; 12:157. [PMID: 33092652 PMCID: PMC7579899 DOI: 10.1186/s13148-020-00951-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Tobacco smoking is a well-known modifiable risk factor for many chronic diseases, including cardiovascular disease (CVD). One of the proposed underlying mechanism linking smoking to disease is via epigenetic modifications, which could affect the expression of disease-associated genes. Here, we conducted a three-way association study to identify the relationship between smoking-related changes in DNA methylation and gene expression and their associations with cardio-metabolic traits. RESULTS We selected 2549 CpG sites and 443 gene expression probes associated with current versus never smokers, from the largest epigenome-wide association study and transcriptome-wide association study to date. We examined three-way associations, including CpG versus gene expression, cardio-metabolic trait versus CpG, and cardio-metabolic trait versus gene expression, in the Rotterdam study. Subsequently, we replicated our findings in The Cooperative Health Research in the Region of Augsburg (KORA) study. After correction for multiple testing, we identified both cis- and trans-expression quantitative trait methylation (eQTM) associations in blood. Specifically, we found 1224 smoking-related CpGs associated with at least one of the 443 gene expression probes, and 200 smoking-related gene expression probes to be associated with at least one of the 2549 CpGs. Out of these, 109 CpGs and 27 genes were associated with at least one cardio-metabolic trait in the Rotterdam Study. We were able to replicate the associations with cardio-metabolic traits of 26 CpGs and 19 genes in the KORA study. Furthermore, we identified a three-way association of triglycerides with two CpGs and two genes (GZMA; CLDND1), and BMI with six CpGs and two genes (PID1; LRRN3). Finally, our results revealed the mediation effect of cg03636183 (F2RL3), cg06096336 (PSMD1), cg13708645 (KDM2B), and cg17287155 (AHRR) within the association between smoking and LRRN3 expression. CONCLUSIONS Our study indicates that smoking-related changes in DNA methylation and gene expression are associated with cardio-metabolic risk factors. These findings may provide additional insights into the molecular mechanisms linking smoking to the development of CVD.
Collapse
|
17
|
Dai N. The Diverse Functions of IMP2/IGF2BP2 in Metabolism. Trends Endocrinol Metab 2020; 31:670-679. [PMID: 32586768 DOI: 10.1016/j.tem.2020.05.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/28/2020] [Accepted: 05/26/2020] [Indexed: 12/13/2022]
Abstract
The human insulin-like growth factor 2 (IGF2) mRNA binding protein family (IMPs/IGF2BPs) is involved in a spectrum of biological processes, including development, tumorigenesis, and stemness. IMPs play a major role in post-transcriptional regulation of RNAs through the ribonucleoprotein complex (RNP). They have emerged as direct mammalian target of rapamycin (mTOR) substrates that coordinate nutrient stimulation and RNA life cycle control. IMP2 is a human type 2 diabetes (T2D) gene associated with impaired insulin secretion. Recently, using murine models, the substantial progress in understanding disease mechanisms has highlighted the significance of IMP2 in metabolism. This new knowledge may have the potential for therapeutic benefit.
Collapse
Affiliation(s)
- Ning Dai
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
18
|
Abuhendi N, Qush A, Naji F, Abunada H, Al Buainain R, Shi Z, Zayed H. Genetic polymorphisms associated with type 2 diabetes in the Arab world: A systematic review and meta-analysis. Diabetes Res Clin Pract 2019; 151:198-208. [PMID: 30954515 DOI: 10.1016/j.diabres.2019.03.037] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 03/27/2019] [Indexed: 02/07/2023]
Abstract
AIMS T2DM reach epidemic levels in the Arab countries. In this study, we aimed to perform a systematic review and meta-analysis to underline the susceptibility genetic profile of Arab patients with T2DM that result from SNPs. METHODS We searched four literature databases (PubMed, Scopus, Science Direct and Web of Science) through January 2019. We included all SNPs in candidate genes with an OR > 1 that were associated with T2DM among Arab patients with T2DM. Statistical programs such as software Review Manager (Version 5.02) and STATA (Version 15.1) were used. The pooled odds ratios (ORs) and 95% confidence intervals (CIs) were calculated with a random effects model or a fixed effect model depending on the heterogeneity among studies. I2 statistics and Egger's tests were performed to assess heterogeneity and publication bias. RESULTS Out of 2245 studies, 47 were used for meta-analysis. We captured 31,307 cases and 26,464 controls in which we collected 71 SNPs in 32 genes. A pooled meta-analysis demonstrated 24-69% increase in T2DM risk. Among the 57 SNPs (in 32 genes) that were not included in the meta-analysis, the OR for diabetes ranged from 1.02 to 5.10, with a median of 1.38 (interquartile range 1.33-2.09). Ten studies examined the association between the TCF7L2 polymorphism rs7903146 and T2DM, leading to an aggregated OR of 1.34 (95%CI 1.27-1.41). CONCLUSION The genetic profile that confer susceptibility to T2DM in Arab patients is diverse. This study may serve as a platform for designing a gene panel for testing the susceptibility to T2DM.
Collapse
Affiliation(s)
- Najat Abuhendi
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha, Qatar
| | - Abeer Qush
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha, Qatar
| | - Fozieh Naji
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha, Qatar
| | - Hanan Abunada
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha, Qatar
| | - Reeham Al Buainain
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha, Qatar
| | - Zumin Shi
- Department of Nutrition, College of Health Sciences, Qatar University, Doha, Qatar
| | - Hatem Zayed
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha, Qatar.
| |
Collapse
|
19
|
Reddy BM, Pranavchand R, Latheef SAA. Overview of genomics and post-genomics research on type 2 diabetes mellitus: Future perspectives and a framework for further studies. J Biosci 2019; 44:21. [PMID: 30837372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
In this review, we briefly outlined salient features of pathophysiology and results of the genetic association studies hitherto conducted on type 2 diabetes. Primarily focusing on the current status of genomic research, we briefly discussed the limited progress made during the post-genomic era and tried to identify the limitations of the post-genomic research strategies. We suggested reanalysis of the existing genomic data through advanced statistical and computational methods and recommended integrated genomics-metabolomics approaches for future studies to facilitate understanding of the gene-environment interactions in the manifestation of the disease. We also propose a framework for research that may be apt for determining the effects of urbanization and changing lifestyles in the manifestation of complex genetic disorders like type 2 diabetes in the Indian populations and offset the confounding effects of both genetic and environmental factors in the natural way.
Collapse
|
20
|
Overview of genomics and post-genomics research on type 2 diabetes mellitus: Future perspectives and a framework for further studies. J Biosci 2019. [DOI: 10.1007/s12038-018-9818-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
21
|
Wheeler E, Marenne G, Barroso I. Genetic aetiology of glycaemic traits: approaches and insights. Hum Mol Genet 2017; 26:R172-R184. [PMID: 28977447 PMCID: PMC5886471 DOI: 10.1093/hmg/ddx293] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 07/18/2017] [Accepted: 07/21/2017] [Indexed: 12/17/2022] Open
Abstract
Glycaemic traits such as fasting and post-challenge glucose and insulin measures, as well as glycated haemoglobin (HbA1c), are used to diagnose and monitor diabetes. These traits are risk factors for cardiovascular disease even below the diabetic threshold, and their study can additionally yield insights into the pathophysiology of type 2 diabetes. To date, a diverse set of genetic approaches have led to the discovery of over 97 loci influencing glycaemic traits. In this review, we will focus on recent advances in the genetic aetiology of glycaemic traits, and the resulting biological insights. We will provide a brief overview of results ranging from common, to low- and rare-frequency variant-trait association studies, studies leveraging the diversity across populations, and studies harnessing the power of genetic and genomic approaches to gain insights into the biological underpinnings of these traits.
Collapse
Affiliation(s)
- Eleanor Wheeler
- Department of Human Genetics, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Gaëlle Marenne
- Department of Human Genetics, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Inês Barroso
- Department of Human Genetics, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridge CB10 1SA, UK
- Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK
| |
Collapse
|
22
|
Ikram MA, Brusselle GGO, Murad SD, van Duijn CM, Franco OH, Goedegebure A, Klaver CCW, Nijsten TEC, Peeters RP, Stricker BH, Tiemeier H, Uitterlinden AG, Vernooij MW, Hofman A. The Rotterdam Study: 2018 update on objectives, design and main results. Eur J Epidemiol 2017; 32:807-850. [PMID: 29064009 PMCID: PMC5662692 DOI: 10.1007/s10654-017-0321-4] [Citation(s) in RCA: 351] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 10/06/2017] [Indexed: 02/07/2023]
Abstract
The Rotterdam Study is a prospective cohort study ongoing since 1990 in the city of Rotterdam in The Netherlands. The study targets cardiovascular, endocrine, hepatic, neurological, ophthalmic, psychiatric, dermatological, otolaryngological, locomotor, and respiratory diseases. As of 2008, 14,926 subjects aged 45 years or over comprise the Rotterdam Study cohort. Since 2016, the cohort is being expanded by persons aged 40 years and over. The findings of the Rotterdam Study have been presented in over 1500 research articles and reports (see www.erasmus-epidemiology.nl/rotterdamstudy ). This article gives the rationale of the study and its design. It also presents a summary of the major findings and an update of the objectives and methods.
Collapse
Affiliation(s)
- M Arfan Ikram
- Department of Epidemiology, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Guy G O Brusselle
- Department of Epidemiology, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
- Department of Respiratory Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Sarwa Darwish Murad
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Gastro-Enterology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Oscar H Franco
- Department of Epidemiology, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
- Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - André Goedegebure
- Department of Otolaryngology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Caroline C W Klaver
- Department of Epidemiology, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Tamar E C Nijsten
- Department of Dermatology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Robin P Peeters
- Department of Epidemiology, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Bruno H Stricker
- Department of Epidemiology, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Henning Tiemeier
- Department of Epidemiology, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - André G Uitterlinden
- Department of Epidemiology, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Meike W Vernooij
- Department of Epidemiology, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
23
|
Dorajoo R, Ali Y, Tay VSY, Kang J, Samydurai S, Liu J, Boehm BO. Single-cell transcriptomics of East-Asian pancreatic islets cells. Sci Rep 2017; 7:5024. [PMID: 28694456 PMCID: PMC5504042 DOI: 10.1038/s41598-017-05266-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 05/25/2017] [Indexed: 12/16/2022] Open
Abstract
Single-cell RNA-seq (scRNA-seq) of pancreatic islets have reported on α- and β-cell gene expression in mice and subjects of predominantly European ancestry. We aimed to assess these findings in East-Asian islet-cells. 448 islet-cells were captured from three East-Asian non-diabetic subjects for scRNA-seq. Hierarchical clustering using pancreatic cell lineage genes was used to assign cells into cell-types. Differentially expressed transcripts between α- and β-cells were detected using ANOVA and in silico replications of mouse and human islet cell genes were performed. We identified 118 α, 105 β, 6 δ endocrine cells and 47 exocrine cells. Besides INS and GCG, 26 genes showed differential expression between α- and β-cells. 10 genes showed concordant expression as reported in rodents, while FAM46A was significantly discordant. Comparing our East-Asian data with data from primarily European subjects, we replicated several genes implicated in nuclear receptor activations, acute phase response pathway, glutaryl-CoA/tryptophan degradations and EIF2/AMPK/mTOR signaling. Additionally, we identified protein ubiquitination to be associated among East-Asian β-cells. We report on East-Asian α- and β-cell gene signatures and substantiate several genes/pathways. We identify expression signatures in East-Asian β-cells that perhaps reflects increased susceptibility to cell-death and warrants future validations to fully appreciate their role in East-Asian diabetes pathogenesis.
Collapse
Affiliation(s)
- Rajkumar Dorajoo
- Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore
| | - Yusuf Ali
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.,Singapore Eye Research Institute, The Academia, Singapore, Singapore
| | - Vanessa S Y Tay
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Jonathan Kang
- Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore
| | - Sudhagar Samydurai
- Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore
| | - Jianjun Liu
- Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore. .,Singapore Eye Research Institute, The Academia, Singapore, Singapore. .,Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore. .,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Bernhard O Boehm
- Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore. .,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore. .,Department of Endocrinology, Tan Tock Seng Hospital, Singapore, Singapore. .,Imperial College London, London, UK.
| |
Collapse
|