1
|
Zannas AS. Cell Type-Level Epigenetics at the Frontier of Atherosclerosis Research. Circulation 2025; 151:1409-1411. [PMID: 40354452 PMCID: PMC12074577 DOI: 10.1161/circulationaha.125.074275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Affiliation(s)
- Anthony S Zannas
- Departments of Psychiatry and Genetics, University of North Carolina at Chapel Hill
| |
Collapse
|
2
|
Zhao JD, Fang ZH. Proteomic Analysis of the Effects of Shenzhu Tiaopi Granules on Model Rats with Type 2 Diabetes Mellitus. Diabetes Metab Syndr Obes 2025; 18:583-599. [PMID: 40026899 PMCID: PMC11871873 DOI: 10.2147/dmso.s493036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 01/22/2025] [Indexed: 03/05/2025] Open
Abstract
Background Shenzhu Tiaopi granule (STG) has antidiabetic functions. Data-independent acquisition proteomic technology is an integral part of systems biology. Herein, proteomics was used to analyse the effects of STG on type 2 diabetes mellitus (T2DM) and the mechanism by which STG normalizes glucose metabolism. Methods Goto-Kakizaki (GK) T2DM model (Mod) rats, aged 15-16 weeks and with a fasting blood glucose (FBG) level of ≥11.1 mmol/L, were treated with metformin or STG for 12 weeks. Wistar rats aged 15-16 weeks were included in the control (Con) group. Body weight, FBG, total cholesterol (TC), total triglyceride (TG) levels and low-density lipoprotein (LDL-C) levels were measured, and pathological observation, Western blot analysis and data-independent acquisition proteomics of the liver were performed. Results Significant differences in FBG, TC, TG, LDL-C (p < 0.01) and pathological liver morphology were observed between the Mod group and Con group, whereas both metformin and STG normalized the glucose and lipid metabolism indicators (p < 0.05 or p < 0.01). In total, 5856 proteins were identified via proteomic analysis, 97 of which were significantly differentially expressed in the liver and affected fatty acid metabolism, unsaturated fatty acid biosynthesis, the peroxisome proliferator-activated receptor (PPAR) signalling pathway, pyruvate metabolism, and terpenoid backbone biosynthesis. Screening identified 10 target proteins, including perilipin-2 (Plin2), pyruvate dehydrogenase kinase 4, farnesyl diphosphate synthase (Fdps) and farnesyl-diphosphate farnesyltransferase 1. Among these proteins, the key proteins were Plin2 and Fdps, which were found to be associated with the PPAR signalling pathway and terpenoid backbone biosynthesis via relationship networks. Plin2 and Fdps are closely related to hyperglycaemia. STG can downregulate Plin2 and upregulate Fdps (p < 0.01). Conclusion STG ameliorated hyperglycaemia by significantly altering the expression of different proteins, especially Fdps and Plin2, in the livers of GK rats. These findings may reveal the potential of traditional Chinese medicine for treating T2DM.
Collapse
Affiliation(s)
- Jin-Dong Zhao
- Department of Endocrinology Two, the First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui Province, 230031, People’s Republic of China
- Center for Xin’an Medicine and Modernization of Traditional Chinese Medicine of IHM, the First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui Province, 230012, People’s Republic of China
- Diabetes Institute, Anhui Academy Chinese Medicine, Hefei, 230012, People’s Republic of China
| | - Zhao-Hui Fang
- Department of Endocrinology Two, the First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui Province, 230031, People’s Republic of China
- Center for Xin’an Medicine and Modernization of Traditional Chinese Medicine of IHM, the First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui Province, 230012, People’s Republic of China
- Diabetes Institute, Anhui Academy Chinese Medicine, Hefei, 230012, People’s Republic of China
| |
Collapse
|
3
|
Bai X, Zhang L, Ji X, Chen K, Di X, Zheng F, Bai P, Niu W. Long-term weight change, incident cardiovascular disease and all-cause mortality among diabetic adults. Diabetes Res Clin Pract 2025; 219:111946. [PMID: 39631677 DOI: 10.1016/j.diabres.2024.111946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
We aimed to explore the impact of weight change from young to middle adulthood on incident cardiovascular disease (CVD) and all-cause mortality in US diabetic adults. All study subjects aged 40-79 years were from the US National Health and Nutrition Examination Survey (NHANES) 1988-2018, and they were non-pregnant and had complete data on self-reported weight at age 25 and 10 years before baseline enrollment over average 29.4 years. CVD events occurring from 10 years ago to baseline enrollment were recorded. Relative to stable non-obesity group, the strongest association was noted for the weight-losing group, followed by the stable-obesity group and weight-gaining group over following 10 years. Referring to the stable-obesity group identified significance for the stable non-obesity group. If total population had maintained non-obese from young to middle adulthood, 12 % of CVD cases could have been averted. Relative to the stable non-obesity group, subjects who maintained obese between young and middle adulthood had an increased risk of all-cause mortality. Our findings indicated that the risk for incident CVD and all-cause mortality was potentially reinforced in diabetic adults who were obese at age 25 but non-obese at midlife and who remained stable obese vis-à-vis those with stable non-obesity.
Collapse
Affiliation(s)
- Xue Bai
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing 100029, China
| | - Lei Zhang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Xiaoxiao Ji
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Kening Chen
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Xueshi Di
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Fangjieyi Zheng
- Center for Evidence-Based Medicine, Capital Institute of Pediatrics, Beijing 100020, China
| | - Peng Bai
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing 100029, China.
| | - Wenquan Niu
- Center for Evidence-Based Medicine, Capital Institute of Pediatrics, Beijing 100020, China.
| |
Collapse
|
4
|
Liu Q, Yu F, Zhou Q, Zhou H, Lu H, Luo J, Gan S. Chinese Visceral Adipose Index is Associated with Arterial Stiffness in Type 2 Diabetes Patients: A Cross-Sectional Study. Int J Gen Med 2024; 17:5681-5696. [PMID: 39650791 PMCID: PMC11622680 DOI: 10.2147/ijgm.s489853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/21/2024] [Indexed: 12/11/2024] Open
Abstract
Purpose The purpose of this study is to investigate the association between Chinese Visceral Adipose Index(CVAI) and brachial-ankle pulse wave velocity (baPWV) in type 2 diabetes (T2D) patients, in order to provide scientific evidence for the prevention and treatment of macrovascular complications in T2D. Patients and Methods This research adopts the cross-sectional study design. Anthropometric assessment, baPWV assessment and biochemical assessment were performed in 2906 T2D patients. CVAI was calculated from the combination of triglycerides, age, high-density lipoprotein cholesterol, waist circumference, body mass index. Multivariate regression analysis and generalized additive model were used to analyze the association between bapwv and CVAI. Subgroup analysis and interaction analysis were used to analyze the influencing factors. Results After adjustment for covariates, baPWV tended to increase in the quartiles of CVAI. In males, As CVAI increases by a unit, baPWV was increased by 0.28 cm/s (95% CI: -0.05, 0.61 P=0.0934). In females, As CVAI increases by a unit, baPWV was increased by 1.60 cm/s (95% CI: 1.07, 2.14). A non-linear connection in males and a linear connection in females between the CVAI and baPWV was revealed. In males, we found that CVAI interacted with baPWV in different duration of diabetes (P interaction = 0.0052), alcohol consumption status (P interaction = 0.0375). In females, CVAI interacted with baPWV in glycated hemoglobin (P interaction = 0.0003), systolic blood pressure (P interaction = 0.0001), diastolic blood pressure (P interaction<0.0001), duration of diabetes (P interaction = 0.0014), the use or non-use of glucose-lowering drugs (P interaction = 0.0006), the use or non-use antihypertensive drugs (P interaction = 0.0004), females' menopausal status (P interaction = 0.0012). Conclusion The relationship between CVAI and baPWV in T2D patients is positively non-linear in males and linear in females. In all subjects, this relationship was influenced by diabetes duration. In males, drinking status affected this relationship, and in females, It is influenced by blood pressure, glycemic control, menopausal status, and the use of glucose-lowering and hypotensive drug, highlighting the complex interplay between visceral fat and arterial stiffness in the T2D population.
Collapse
Affiliation(s)
- Qin Liu
- Department of Endocrinology, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, People’s Republic of China
| | - Fang Yu
- Department of Endocrinology, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, People’s Republic of China
| | - Quan Zhou
- Department of Science and Education, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, People’s Republic of China
| | - Haifeng Zhou
- Department of Endocrinology, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, People’s Republic of China
| | - Huilin Lu
- Department of Endocrinology, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, People’s Republic of China
| | - Jian Luo
- Department of Endocrinology, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, People’s Republic of China
| | - Shenglian Gan
- Department of Endocrinology, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, People’s Republic of China
| |
Collapse
|
5
|
Chen M, Yang Y, Chen S, He Z, Du L. Targeting squalene epoxidase in the treatment of metabolic-related diseases: current research and future directions. PeerJ 2024; 12:e18522. [PMID: 39588004 PMCID: PMC11587872 DOI: 10.7717/peerj.18522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/23/2024] [Indexed: 11/27/2024] Open
Abstract
Metabolic-related diseases are chronic diseases caused by multiple factors, such as genetics and the environment. These diseases are difficult to cure and seriously affect human health. Squalene epoxidase (SQLE), the second rate-limiting enzyme in cholesterol synthesis, plays an important role in cholesterol synthesis and alters the gut microbiota and tumor immunity. Research has shown that SQLE is expressed in many tissues and organs and is involved in the occurrence and development of various metabolic-related diseases, such as cancer, nonalcoholic fatty liver disease, diabetes mellitus, and obesity. SQLE inhibitors, such as terbinafine, NB598, natural compounds, and their derivatives, can effectively ameliorate fungal infections, nonalcoholic fatty liver disease, and cancer. In this review, we provide an overview of recent research progress on the role of SQLE in metabolic-related diseases. Further research on the regulation of SQLE expression is highly important for developing drugs for the treatment of metabolic-related diseases with good pharmacological activity.
Collapse
Affiliation(s)
- Mingzhu Chen
- School of Basic Medical Sciences, Chengdu University of Chinese Medicine, Chengdu, Sichuan Province, China
| | - Yongqi Yang
- Harbin Medical University, Department of Pharmacology, College of Pharmacy, Harbin, Heilongjiang Province, China
| | - Shiting Chen
- School of Basic Medical Sciences, Chengdu University of Chinese Medicine, Chengdu, Sichuan Province, China
| | - Zhigang He
- School of Basic Medical Sciences, Chengdu University of Chinese Medicine, Chengdu, Sichuan Province, China
| | - Lian Du
- School of Basic Medical Sciences, Chengdu University of Chinese Medicine, Chengdu, Sichuan Province, China
| |
Collapse
|
6
|
Chen J, Rao H, Zheng X. Identification of novel targets associated with cholesterol metabolism in nonalcoholic fatty liver disease: a comprehensive study using Mendelian randomization combined with transcriptome analysis. Front Genet 2024; 15:1464865. [PMID: 39359475 PMCID: PMC11445148 DOI: 10.3389/fgene.2024.1464865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/06/2024] [Indexed: 10/04/2024] Open
Abstract
Background There is limited research on cholesterol metabolism-related genes (CM-RGs) in non-alcoholic fatty liver disease (NAFLD), despite hypercholesterolemia being a recognized risk factor. The role of CM-RGs in NAFLD remains unclear. Methods The differentially expressed genes (DEGs) between NAFLD and control were acquired by differential expression analysis. The differentially expressed genes associated with cholesterol metabolism (DE-CM-RGs) were identified and functional enrichment analyses were performed. Protein-protein interaction network analysis and a two-sample Mendelian randomization study were utilized for identifying hub genes. Nomogram model, competing endogenous RNA and messenger RNA-drug networks were established. In addition, immunoinfiltration analysis was performed. Results We identified four hub genes (MVK, HMGCS1, TM7SF2, and FDPS) linked to NAFLD risk. MVK and TM7SF2 were protective factors, HMGCS1 and FDPS were risk factors for NAFLD. The area under the curve values of nomograms in GSE135251 and GSE126848 were 0.79 and 0.848, respectively. The gene set enrichment analysis indicated that hub genes participated in calcium signaling pathways and biosynthesis of unsaturated fatty acids. NAFLD patients showed increased CD56dim NK cells and Th17. Tretinoin, alendronate, zoledronic acid, and quercetin are potential target agents in NAFLD. Conclusion Our study has linked cholesterol metabolism genes (MVK, HMGCS1, TM7SF2, and FDPS) to NAFLD, providing a promising diagnostic framework, identifying treatment targets, and offering novel perspectives into its mechanisms.
Collapse
Affiliation(s)
- Juan Chen
- Department of Gastroenterology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Huajing Rao
- Emergency Internal Medicine, Affiliated Fuzhou First Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Xiaoling Zheng
- Department of Endoscopy, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| |
Collapse
|
7
|
Kang H, Song J, Cheng Y. HDL regulates the risk of cardiometabolic and inflammatory-related diseases: Focusing on cholesterol efflux capacity. Int Immunopharmacol 2024; 138:112622. [PMID: 38971111 DOI: 10.1016/j.intimp.2024.112622] [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: 06/08/2024] [Revised: 06/29/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
Dyslipidemia, characterized by higher serum concentrations of low-density lipoprotein cholesterol (LDL-C), very low-density lipoprotein cholesterol (VLDL-C), triglyceride (TG), and lower serum concentrations of high-density lipoprotein cholesterol (HDL-C), is confirmed as a hallmark of cardiovascular diseases (CVD), posing serious risks to the future health of humans. Aside from the role of HDL-C concentrations, the capacity of cholesterol efflux to HDL is being identified as an enssential messurement for the dyslipidemic morbidity. Through inducing the progression of reverse cholesterol transport (RCT), the HDL-related cholesterol efflux plays a vital role in atherosclerotic plaque formation. In addition, increasing results demonstrated that the relationships between cholesterol efflux and cardiovascular events might be influenced by multiple factors, such as atherosclerosis, diabetes, and, inflammatory diseases. These risk factors could affect the intracellular composition of HDL, which might subsqently influence the cholesterol efflux process induced by HDL particle. In the present comprehensive article, we summarize the latest findings which described the modulatory roles of HDL in cardiometabolic disorders and inflammatory related diseases, focusing on its capacity in mediating cholesterol efflux. Moreover, the potential mechanisms whereby HDL regulate the risk of cardiometabolic disorders or inflammatory related diseases, at least partly, via cholesterol efflux pathway, are also well-listed.
Collapse
Affiliation(s)
- Huiyuan Kang
- Department of Cardiology, the Xiamen Cardiovascular Hospital of Xiamen University, Xiamen, Fujian, China
| | - Jingjin Song
- Department of Cardiology, the Xiamen Cardiovascular Hospital of Xiamen University, Xiamen, Fujian, China
| | - Ye Cheng
- Department of Cardiology, the Xiamen Cardiovascular Hospital of Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
8
|
Xing X, Xu P, Xing X, Xu Z, Huang Z, Li Z, Li X, Xiao Y. Effects of ADSC-Derived Exosome LRRC75A-AS1 on Anti-inflammatory Function After SCI. Appl Biochem Biotechnol 2024; 196:5920-5935. [PMID: 38165592 DOI: 10.1007/s12010-023-04836-9] [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] [Accepted: 12/19/2023] [Indexed: 01/04/2024]
Abstract
Spinal cord injury (SCI) is a highly debilitating disorder of the central nervous system that can severely impact an affected patient's quality of life. This study aimed to examine how adipose-derived mesenchymal stem cell exosomes (ADSC-exos) can be used to treat spinal cord injury. We analysed differentially expressed mRNAs in SCI using bioinformatics data, gene expression profiles in inflammatory cell models, RT-qPCR and WB. Apoptosis was detected with flow cytometry. Starbase provides the control mechanism for FDFT1. Target interactions were detected with dual-luciferase reporter and RIP assays. Exosomes were isolated from adipose tissue-derived mesenchymal stem cells and subsequently characterized with western blot analysis, transmission electron microscopy and nanoparticle tracking analysis. By analysing the GSE102964 database, we found that FDFT1 was significantly downregulated as SCI progressed. Overexpression of FDFT1 can significantly reverse the inflammatory response and apoptosis of BV2 cells induced by hemin. Mechanically, ADSC-exos can affect the expression of FDFT1 through the ceRNA mechanism mediated by LRRC75A-AS1 and in an RBP-dependent manner mediated by IGF2BP2. The overexpression of LRRC75A-AS1 significantly enhances BV2 apoptosis and can be reversed by FDFT1 knockdown. ADSC-exos LRRC75A-AS1 inhibits inflammation and reduces SCI by increasing the expression and stability of FDFT1 mRNA in a ceRNA and RBP-dependent manner.
Collapse
Affiliation(s)
- Xiaohui Xing
- Department of Neurosurgery, Liaocheng People's Hospital, No. 67 Dongchang West Road, Liaocheng, 252000, China
| | - Peng Xu
- Department of Neurosurgery, Liaocheng People's Hospital, No. 67 Dongchang West Road, Liaocheng, 252000, China
| | - Xiaoyang Xing
- Department of Laboratory, Liaocheng Maternal and Child Health Care Hospital, No.56, Changjiang Road, Liaocheng, 252000, Shandong, China
| | - Zhentao Xu
- Department of Neurosurgery, Liaocheng People's Hospital, No. 67 Dongchang West Road, Liaocheng, 252000, China
| | - Zhen Huang
- Department of Neurosurgery, Liaocheng People's Hospital, No. 67 Dongchang West Road, Liaocheng, 252000, China
| | - Zhongchen Li
- Department of Neurosurgery, Liaocheng People's Hospital, No. 67 Dongchang West Road, Liaocheng, 252000, China
| | - Xueyuan Li
- Department of Neurosurgery, Liaocheng People's Hospital, No. 67 Dongchang West Road, Liaocheng, 252000, China.
| | - Yilei Xiao
- Department of Neurosurgery, Liaocheng People's Hospital, No. 67 Dongchang West Road, Liaocheng, 252000, China.
| |
Collapse
|
9
|
Warner B, Ratner E, Datta A, Lendasse A. A systematic review of phenotypic and epigenetic clocks used for aging and mortality quantification in humans. Aging (Albany NY) 2024; 16:12414-12427. [PMID: 39215995 PMCID: PMC11424583 DOI: 10.18632/aging.206098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 07/15/2024] [Indexed: 09/04/2024]
Abstract
Aging is the leading driver of disease in humans and has profound impacts on mortality. Biological clocks are used to measure the aging process in the hopes of identifying possible interventions. Biological clocks may be categorized as phenotypic or epigenetic, where phenotypic clocks use easily measurable clinical biomarkers and epigenetic clocks use cellular methylation data. In recent years, methylation clocks have attained phenomenal performance when predicting chronological age and have been linked to various age-related diseases. Additionally, phenotypic clocks have been proven to be able to predict mortality better than chronological age, providing intracellular insights into the aging process. This review aimed to systematically survey all proposed epigenetic and phenotypic clocks to date, excluding mitotic clocks (i.e., cancer risk clocks) and those that were modeled using non-human samples. We reported the predictive performance of 33 clocks and outlined the statistical or machine learning techniques used. We also reported the most influential clinical measurements used in the included phenotypic clocks. Our findings provide a systematic reporting of the last decade of biological clock research and indicate possible avenues for future research.
Collapse
Affiliation(s)
| | | | | | - Amaury Lendasse
- Department of IST, University of Houston, Houston, TX 77004, USA
- Department of Engineering Management and Systems Engineering, Missouri University of Science and Technology, Rolla, MO 65409, USA
| |
Collapse
|
10
|
da Silva Rodrigues Marçal E, Borges JB, Bastos GM, Crespo Hirata TD, de Oliveira VF, Gonçalves RM, Faludi AA, Dias França JI, de Oliveira Silva DV, Malaquias VB, Luchessi AD, Silbiger VN, Nakazone MA, Carmo TS, Silva Souza DR, Sampaio MF, Crespo Hirata RD, Hirata MH. Methylation status of LDLR, PCSK9 and LDLRAP1 is associated with cardiovascular events in familial hypercholesterolemia. Epigenomics 2024; 16:809-820. [PMID: 38884343 PMCID: PMC11370914 DOI: 10.1080/17501911.2024.2351792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 04/27/2024] [Indexed: 06/18/2024] Open
Abstract
Aim: Methylation of LDLR, PCSK9 and LDLRAP1 CpG sites was assessed in patients with familial hypercholesterolemia (FH). Methods: DNA methylation of was analyzed by pyrosequencing in 131 FH patients and 23 normolipidemic (NL) subjects.Results: LDLR, PCSK9 and LDLRP1 methylation was similar between FH patients positive (MD) and negative (non-MD) for pathogenic variants in FH-related genes. LDLR and PCSK9 methylation was higher in MD and non-MD groups than NL subjects (p < 0.05). LDLR, PCSK9 and LDLRAP1 methylation profiles were associated with clinical manifestations and cardiovascular events in FH patients (p < 0.05).Conclusion: Differential methylation of LDLR, PCSK9 and LDLRAP1 is associated with hypercholesterolemia and cardiovascular events. This methylation profile maybe useful as a biomarker and contribute to the management of FH.
Collapse
Affiliation(s)
- Elisangela da Silva Rodrigues Marçal
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, 05508-000, Brazil
- Laboratory of Molecular Research in Cardiology, Institute of Cardiology Dante Pazzanese, Sao Paulo, 04012-909, Brazil
| | - Jéssica Bassani Borges
- Department of Research, Hospital Beneficiencia Portuguesa de Sao Paulo, Sao Paulo, 01323-001, Brazil
| | - Gisele Medeiros Bastos
- Department of Research, Hospital Beneficiencia Portuguesa de Sao Paulo, Sao Paulo, 01323-001, Brazil
| | - Thiago Dominguez Crespo Hirata
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, 05508-000, Brazil
| | - Victor Fernandes de Oliveira
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, 05508-000, Brazil
| | | | - Andre Arpad Faludi
- Medical Clinic Division, Institute of Cardiology Dante Pazzanese, Sao Paulo, 04012-909, Brazil
| | - João Italo Dias França
- Center for Clinical Trials & Pharmacovigilance, Butantan Institute, Sao Paulo, 05585-000, Brazil
| | - Daiana Vitor de Oliveira Silva
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, 05508-000, Brazil
| | - Vanessa Barbosa Malaquias
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, 05508-000, Brazil
| | - Andre Ducati Luchessi
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, Federal University of Rio Grande do Norte, Natal, 59012-570, Brazil
- Graduate Program in Pharmaceutical Sciences, Federal University of Rio Grande do Norte, Natal, 59012-570, Brazil
| | - Vivian Nogueira Silbiger
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, Federal University of Rio Grande do Norte, Natal, 59012-570, Brazil
- Graduate Program in Pharmaceutical Sciences, Federal University of Rio Grande do Norte, Natal, 59012-570, Brazil
| | - Marcelo Arruda Nakazone
- Department of Cardiology & Cardiovascular Surgery, Sao Jose do Rio Preto Medical School, Sao Jose do Rio Preto, 15090-000, Brazil
| | - Tayanne Silva Carmo
- Department of Biochemistry & Molecular Biology, Sao Jose do Rio Preto Medical School, Sao Jose do Rio Preto, 15090-000, Brazil
| | - Dorotéia Rossi Silva Souza
- Department of Biochemistry & Molecular Biology, Sao Jose do Rio Preto Medical School, Sao Jose do Rio Preto, 15090-000, Brazil
| | - Marcelo Ferraz Sampaio
- Department of Cardiology, Hospital Beneficencia Portuguesa de Sao Paulo, Sao Paulo, 01323-001, Brazil
| | - Rosario Dominguez Crespo Hirata
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, 05508-000, Brazil
| | - Mario Hiroyuki Hirata
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, 05508-000, Brazil
| |
Collapse
|
11
|
Lewis KA, Stroebel BM, Zhang L, Aouizerat B, Mattis AN, Flowers E. MicroRNAs Associated with Metformin Treatment in the Diabetes Prevention Program. Int J Mol Sci 2024; 25:5684. [PMID: 38891870 PMCID: PMC11172132 DOI: 10.3390/ijms25115684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/16/2024] [Accepted: 05/18/2024] [Indexed: 06/21/2024] Open
Abstract
The Diabetes Prevention Program (DPP) randomized controlled trial demonstrated that metformin treatment reduced progression to type 2 diabetes (T2D) by 31% compared to placebo in adults with prediabetes. Circulating micro-ribonucleic acids (miRs) are promising biomarkers of T2D risk, but little is known about their associations with metformin regimens for T2D risk reduction. We compared the change in 24 circulating miRs from baseline to 2 years in a subset from DPP metformin intervention (n = 50) and placebo (n = 50) groups using Wilcoxon signed rank tests. Spearman correlations were used to evaluate associations between miR change and baseline clinical characteristics. Multiple linear regression was used to adjust for covariates. The sample was 73% female, 17% Black, 13% Hispanic, and 50 ± 11 years. Participants were obese, normotensive, prediabetic, and dyslipidemic. Change in 12 miR levels from baseline to 2 years was significantly different in the metformin group compared with placebo after adjusting for multiple comparisons: six (let-7c-5p, miR-151a-3p, miR-17-5p, miR-20b-5p, miR-29b-3p, and miR-93-5p) were significantly upregulated and six (miR-130b-3p, miR-22-3p, miR-222-3p, miR-320a-3p, miR-320c, miR-92a-3p) were significantly downregulated in the metformin group. These miRs help to explain how metformin is linked to T2D risk reduction, which may lead to novel biomarkers, therapeutics, and precision health strategies.
Collapse
Affiliation(s)
- Kimberly A. Lewis
- Department of Physiological Nursing, School of Nursing, University of California, 2 Koret Way, San Francisco, CA 94143, USA; (B.M.S.); (E.F.)
| | - Benjamin M. Stroebel
- Department of Physiological Nursing, School of Nursing, University of California, 2 Koret Way, San Francisco, CA 94143, USA; (B.M.S.); (E.F.)
| | - Li Zhang
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA 94143, USA;
| | | | - Aras N. Mattis
- Department of Pathology, University of California, San Francisco, CA 94143, USA;
| | - Elena Flowers
- Department of Physiological Nursing, School of Nursing, University of California, 2 Koret Way, San Francisco, CA 94143, USA; (B.M.S.); (E.F.)
- Institute for Human Genetics, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
12
|
Ding J, Nguyen AT, Lohman K, Hensley MT, Parker D, Hou L, Taylor J, Voora D, Sawyer JK, Boudyguina E, Bancks MP, Bertoni A, Pankow JS, Rotter JI, Goodarzi MO, Tracy RP, Murdoch DM, Duprez D, Rich SS, Psaty BM, Siscovick D, Newgard CB, Herrington D, Hoeschele I, Shea S, Stein JH, Patel M, Post W, Jacobs D, Parks JS, Liu Y. LXR signaling pathways link cholesterol metabolism with risk for prediabetes and diabetes. J Clin Invest 2024; 134:e173278. [PMID: 38747290 PMCID: PMC11093600 DOI: 10.1172/jci173278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 03/20/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUNDPreclinical studies suggest that cholesterol accumulation leads to insulin resistance. We previously reported that alterations in a monocyte cholesterol metabolism transcriptional network (CMTN) - suggestive of cellular cholesterol accumulation - were cross-sectionally associated with obesity and type 2 diabetes (T2D). Here, we sought to determine whether the CMTN alterations independently predict incident prediabetes/T2D risk, and correlate with cellular cholesterol accumulation.METHODSMonocyte mRNA expression of 11 CMTN genes was quantified among 934 Multi-Ethnic Study of Atherosclerosis (MESA) participants free of prediabetes/T2D; cellular cholesterol was measured in a subset of 24 monocyte samples.RESULTSDuring a median 6-year follow-up, lower expression of 3 highly correlated LXR target genes - ABCG1 and ABCA1 (cholesterol efflux) and MYLIP (cholesterol uptake suppression) - and not other CMTN genes, was significantly associated with higher risk of incident prediabetes/T2D. Lower expression of the LXR target genes correlated with higher cellular cholesterol levels (e.g., 47% of variance in cellular total cholesterol explained by ABCG1 expression). Further, adding the LXR target genes to overweight/obesity and other known predictors significantly improved prediction of incident prediabetes/T2D.CONCLUSIONThese data suggest that the aberrant LXR/ABCG1-ABCA1-MYLIP pathway (LAAMP) is a major T2D risk factor and support a potential role for aberrant LAAMP and cellular cholesterol accumulation in diabetogenesis.FUNDINGThe MESA Epigenomics and Transcriptomics Studies were funded by NIH grants 1R01HL101250, 1RF1AG054474, R01HL126477, R01DK101921, and R01HL135009. This work was supported by funding from NIDDK R01DK103531 and NHLBI R01HL119962.
Collapse
Affiliation(s)
- Jingzhong Ding
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | | | - Kurt Lohman
- Department of Medicine, Division of Cardiology, and
| | | | - Daniel Parker
- Department of Medicine, Division of Geriatrics, Duke University, Durham, North Carolina, USA
| | - Li Hou
- Department of Medicine, Division of Cardiology, and
| | - Jackson Taylor
- Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, Ohio, USA
| | - Deepak Voora
- Department of Medicine, Division of Cardiology, and
| | - Janet K. Sawyer
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Elena Boudyguina
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Michael P. Bancks
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Alain Bertoni
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - James S. Pankow
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota, 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, California, USA
| | - Mark O. Goodarzi
- Division of Endocrinology, Diabetes and Metabolism, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Russell P. Tracy
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | - David M. Murdoch
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University, Durham, North Carolina, USA
| | - Daniel Duprez
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology, and Health Systems and Population Health, University of Washington, Seattle, Washington, USA
| | | | - Christopher B. Newgard
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, USA
| | - David Herrington
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Ina Hoeschele
- Fralin Life Sciences Institute, Virginia Tech, Blacksburg, Virginia, USA
| | - Steven Shea
- Department of Medicine, Columbia University, New York, New York, USA
| | - James H. Stein
- School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| | - Manesh Patel
- Department of Medicine, Division of Cardiology, and
| | - Wendy Post
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - David Jacobs
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - John S. Parks
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Yongmei Liu
- Department of Medicine, Division of Cardiology, and
| |
Collapse
|
13
|
Lützhøft DO, Bækgård C, Wimborne E, Straarup EM, Pedersen KM, Swann JR, Pedersen HD, Kristensen K, Morgills L, Nielsen DS, Hansen AK, Bracken MK, Cirera S, Christoffersen BØ. High fat diet is associated with gut microbiota dysbiosis and decreased gut microbial derived metabolites related to metabolic health in young Göttingen Minipigs. PLoS One 2024; 19:e0298602. [PMID: 38427692 PMCID: PMC10906878 DOI: 10.1371/journal.pone.0298602] [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: 07/06/2023] [Accepted: 01/26/2024] [Indexed: 03/03/2024] Open
Abstract
The objectives were 1) to characterize a Göttingen Minipig model of metabolic syndrome regarding its colon microbiota and circulating microbial products, and 2) to assess whether ovariectomized female and castrated male minipigs show similar phenotypes. Twenty-four nine-week-old Göttingen Minipigs were allocated to four groups based on sex and diet: ovariectomized females and castrated males fed either chow or high-fat diet (HFD) for 12 weeks. At study end, body composition and plasma biomarkers were measured, and a mixed meal tolerance test (MMT) and an intravenous glucose tolerance test (IVGTT) were performed. The HFD groups had significantly higher weight gain, fat percentage, fasting plasma insulin and glucagon compared to the chow groups. Homeostatic model assessment of insulin resistance index (HOMA-IR) was increased and glucose effectiveness derived from the IVGTT and Matsuda´s insulin sensitivity index from the MMT were decreased in the HFD groups. The HFD groups displayed dyslipidemia, with significantly increased total-, LDL- and HDL-cholesterol, and decreased HDL/non-HDL cholesterol ratio. The colon microbiota of HFD minipigs clearly differed from the lean controls (GuniFrac distance matrix). The main bacteria families driving this separation were Clostridiaceae, Fibrobacteraceae, Flavobacteriaceae and Porphyromonadaceae. Moreover, the species richness was significantly decreased by HFD. In addition, HFD decreased the circulating level of short chain fatty acids and beneficial microbial metabolites hippuric acid, xanthine and trigonelline, while increasing the level of branched chain amino acids. Six and nine metabolically relevant genes were differentially expressed between chow-fed and HFD-fed animals in liver and omental adipose tissue, respectively. The HFD-fed pigs presented with metabolic syndrome, gut microbial dysbiosis and a marked decrease in healthy gut microbial products and thus displayed marked parallels to human obesity and insulin resistance. HFD-fed Göttingen Minipig therefore represents a relevant animal model for studying host-microbiota interactions. No significant differences between the castrated and ovariectomized minipigs were observed.
Collapse
Affiliation(s)
- Ditte Olsen Lützhøft
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Cecilie Bækgård
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Elizabeth Wimborne
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | | | | | - Jonathan R. Swann
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | | | | | | | - Dennis Sandris Nielsen
- Department of Food Science, Faculty of Science, University of Copenhagen, Frederiksberg C, Denmark
| | - Axel Kornerup Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | | | - Susanna Cirera
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | | |
Collapse
|
14
|
Lewis KA, Stroebel B, Zhang L, Aouizerat B, Mattis A, Flowers E. MicroRNAs Associated with Metformin Treatment in the Diabetes Prevention Program. RESEARCH SQUARE 2024:rs.3.rs-3846347. [PMID: 38313262 PMCID: PMC10836103 DOI: 10.21203/rs.3.rs-3846347/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
The Diabetes Prevention Program (DPP) randomized controlled trial demonstrated that metformin treatment reduced progression to type 2 diabetes (T2D) by 31% compared to placebo in adults with prediabetes. Circulating micro-ribonucleic acids (miRs) are promising biomarkers of T2D risk, but little is known about their associations with metformin regimens for T2D risk reduction. We compared the change in 24 circulating miRs from baseline to 2 years in a subset from DPP metformin intervention (n = 50) and placebo (n = 50) groups using Wilcoxon signed rank tests. Spearman's correlations were used to evaluate associations between miR change and baseline clinical characteristics. Multiple linear regression was used to adjust for covariates. The sample was 73% female, 17% Black, 13% Hispanic, and 50 ± 11 years. Participants were obese, normotensive, prediabetic, and dyslipidemic. Change in 12 miR levels from baseline to 2 years was significantly different in the metformin group compared with placebo after adjusting for multiple comparisons: six (let-7c-5p, miR-151a-3p, miR-17-5p, miR-20b-5p, miR-29b-3p, and miR-93-5p) were significantly upregulated and six (miR-130b-3p, miR-22-3p, miR-222-3p, miR-320a-3p, miR-320c, miR-92a-3p) were significantly downregulated in the metformin group. These miRs help to explain how metformin is linked to T2D risk reduction, which may lead to novel biomarkers, therapeutics, and precision-health strategies.
Collapse
Affiliation(s)
| | | | - Li Zhang
- University of California San Francisco
| | | | | | | |
Collapse
|
15
|
Asam K, Lewis KA, Kober K, Gong X, Kanaya AM, Aouizerat BE, Flowers E. Multi-Tiered Assessment of Gene Expression Provides Evidence for Mechanisms That Underlie Risk for Type 2 Diabetes. Diabetes Metab Syndr Obes 2023; 16:3445-3457. [PMID: 37929060 PMCID: PMC10625391 DOI: 10.2147/dmso.s428572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/25/2023] [Indexed: 11/07/2023] Open
Abstract
Introduction Integrated transcriptome and microRNA differential gene expression (DEG) analyses may help to explain type 2 diabetes (T2D) pathogenesis in at-risk populations. The purpose of this study was to characterize DEG in banked biospecimens from underactive adult participants who responded to a randomized clinical trial measuring the effects of lifestyle interventions on T2D risk factors. DEGs were further examined within the context of annotated biological pathways. Methods Participants (n = 52) in a previously completed clinical trial that assessed a 12-week behavioural intervention for T2D risk reduction were included. Participants who showed >6mg/dL decrease in fasting blood glucose were identified as responders. Gene expression was measured by RNASeq, and overrepresentation analysis within KEGG pathways and weighted gene correlation network analysis (WGCNA) were performed. Results No genes remained significantly differentially expressed after correction for multiple comparisons. One module derived by WGCNA related to body mass index was identified, which contained genes located in KEGG pathways related to known mechanisms underlying risk for T2D as well as pathways related to neurodegeneration and protein misfolding. A network analysis showed indirect connections between genes in this module and islet amyloid polypeptide (IAPP), which has previously been hypothesized as a mechanism for T2D. Discussion We validated prior studies that showed pathways related to metabolism, inflammation/immunity, and endocrine/hormone function are related to risk for T2D. We identified evidence for new potential mechanisms that include protein misfolding. Additional studies are needed to determine whether these are potential therapeutic targets to decrease risk for T2D.
Collapse
Affiliation(s)
- Kesava Asam
- Bluestone Center for Clinical Research, New York University, New York City, NY, USA
| | - Kimberly A Lewis
- Department of Physiological Nursing, University of California, San Francisco, CA, USA
| | - Kord Kober
- Department of Physiological Nursing, University of California, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA, USA
| | - Xingyue Gong
- Department of Physiological Nursing, University of California, San Francisco, CA, USA
| | - Alka M Kanaya
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Bradley E Aouizerat
- Bluestone Center for Clinical Research, New York University, New York City, NY, USA
- Department of Oral and Maxillofacial Surgery, New York University, New York City, NY, USA
| | - Elena Flowers
- Department of Physiological Nursing, University of California, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, CA, USA
| |
Collapse
|
16
|
Cai Y, Zhang S, Chen L, Fu Y. Integrated multi-omics and machine learning approach reveals lipid metabolic biomarkers and signaling in age-related meibomian gland dysfunction. Comput Struct Biotechnol J 2023; 21:4215-4227. [PMID: 37675286 PMCID: PMC10480060 DOI: 10.1016/j.csbj.2023.08.026] [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: 03/11/2023] [Revised: 08/26/2023] [Accepted: 08/26/2023] [Indexed: 09/08/2023] Open
Abstract
Meibomian gland dysfunction (MGD) is a prevalent inflammatory disorder of the ocular surface that significantly impacts patients' vision and quality of life. The underlying mechanism of aging and MGD remains largely uncharacterized. The aim of this work is to investigate lipid metabolic alterations in age-related MGD (ARMGD) through integrated proteomics, lipidomics and machine learning (ML) approach. For this purpose, we collected samples of female mouse meibomian glands (MGs) dissected from eyelids at age two months (n = 9) and two years (n = 9) for proteomic and lipidomic profilings using the liquid chromatography with tandem mass spectrometry (LC-MS/MS) method. To further identify ARMGD-related lipid biomarkers, ML model was established using the least absolute shrinkage and selection operator (LASSO) algorithm. For proteomic profiling, 375 differentially expressed proteins were detected. Functional analyses indicated the leading role of cholesterol biosynthesis in the aging process of MGs. Several proteins were proposed as potential biomarkers, including lanosterol synthase (Lss), 24-dehydrocholesterol reductase (Dhcr24), and farnesyl diphosphate farnesyl transferase 1 (Fdft1). Concomitantly, lipidomic analysis unveiled 47 lipid species that were differentially expressed and clustered into four classes. The most notable age-related alterations involved a decline in cholesteryl esters (ChE) levels and an increase in triradylglycerols (TG) levels, accompanied by significant differences in their lipid unsaturation patterns. Through ML construction, it was confirmed that ChE(26:0), ChE(26:1), and ChE(30:1) represent the most promising diagnostic molecules. The present study identified essential proteins, lipids, and signaling pathways in age-related MGD (ARMGD), providing a reference landscape to facilitate novel strategies for the disease transformation.
Collapse
Affiliation(s)
- Yuchen Cai
- Department of Ophthalmology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Siyi Zhang
- Department of Ophthalmology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Liangbo Chen
- Department of Ophthalmology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Yao Fu
- Department of Ophthalmology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| |
Collapse
|
17
|
Lam S, Lui DTW, Shiu SWM, Wong Y, Tan KCB. Effect of type 2 diabetes on the inducible degrader of LDL receptor. J Lipid Res 2023; 64:100380. [PMID: 37094639 PMCID: PMC10230263 DOI: 10.1016/j.jlr.2023.100380] [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: 06/07/2022] [Revised: 04/12/2023] [Accepted: 04/15/2023] [Indexed: 04/26/2023] Open
Abstract
The inducible degrader of LDL receptor (IDOL) acts as a post-transcriptional degrader of the LDL receptor (LDLR). IDOL is functionally active in the liver and in peripheral tissues. We have evaluated IDOL expression in circulating monocytes in subjects with and without type 2 diabetes and determined whether changes in IDOL expression could affect macrophage function like cytokine production in vitro. One hundred forty individuals with type 2 diabetes and 110 healthy control subjects were recruited. Cellular expression of IDOL and LDLR in peripheral blood CD14+ monocytes was measured by flow cytometry. The expression of intracellular IDOL was lower in individuals with diabetes than control (21.3 ± 4.6 mean fluorescence intensity × 1,000 vs. 23.8 ± 6.2, P < 0.01), and this was accompanied by an increase in cell surface LDLR (5.2 ± 3.0 mean fluorescence intensity × 1,000 vs. 4.3 ± 1.5, P < 0.01), LDL binding, and intracellular lipid (P < 0.01). IDOL expression correlated with HbA1c (r = -0.38, P < 0.01) and serum fibroblast growth factor-21 (FGF21) (r = -0.34, P < 0.01). Multivariable regression analysis, including age, sex, BMI, smoking, HbA1c, and log(FGF21), showed that HbA1c and FGF21 were significant independent determinants of IDOL expression. IDOL knockdown human monocyte-derived macrophages produced higher concentrations of interleukin 1 beta, interleukin 6, and TNFα than control macrophages upon stimulation with lipopolysaccharide (all P < 0.01). In conclusion, the expression of IDOL in CD14+ monocytes was decreased in type 2 diabetes and was associated with glycemia and serum FGF21 concentration.
Collapse
Affiliation(s)
- Sum Lam
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - David Tak Wai Lui
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | | | - Ying Wong
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | | |
Collapse
|
18
|
Covert JD, Grice BA, Thornburg MG, Kaur M, Ryan AP, Tackett L, Bhamidipati T, Stull ND, Kim T, Habegger KM, McClain DA, Brozinick JT, Elmendorf JS. An early, reversible cholesterolgenic etiology of diet-induced insulin resistance. Mol Metab 2023; 72:101715. [PMID: 37019209 PMCID: PMC10114231 DOI: 10.1016/j.molmet.2023.101715] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/27/2023] [Accepted: 03/21/2023] [Indexed: 04/07/2023] Open
Abstract
OBJECTIVE A buildup of skeletal muscle plasma membrane (PM) cholesterol content in mice occurs within 1 week of a Western-style high-fat diet and causes insulin resistance. The mechanism driving this cholesterol accumulation and insulin resistance is not known. Promising cell data implicate that the hexosamine biosynthesis pathway (HBP) triggers a cholesterolgenic response via increasing the transcriptional activity of Sp1. In this study we aimed to determine whether increased HBP/Sp1 activity represented a preventable cause of insulin resistance. METHODS C57BL/6NJ mice were fed either a low-fat (LF, 10% kcal) or high-fat (HF, 45% kcal) diet for 1 week. During this 1-week diet the mice were treated daily with either saline or mithramycin-A (MTM), a specific Sp1/DNA-binding inhibitor. A series of metabolic and tissue analyses were then performed on these mice, as well as on mice with targeted skeletal muscle overexpression of the rate-limiting HBP enzyme glutamine-fructose-6-phosphate-amidotransferase (GFAT) that were maintained on a regular chow diet. RESULTS Saline-treated mice fed this HF diet for 1 week did not have an increase in adiposity, lean mass, or body mass while displaying early insulin resistance. Consistent with an HBP/Sp1 cholesterolgenic response, Sp1 displayed increased O-GlcNAcylation and binding to the HMGCR promoter that increased HMGCR expression in skeletal muscle from saline-treated HF-fed mice. Skeletal muscle from these saline-treated HF-fed mice also showed a resultant elevation of PM cholesterol with an accompanying loss of cortical filamentous actin (F-actin) that is essential for insulin-stimulated glucose transport. Treating these mice daily with MTM during the 1-week HF diet fully prevented the diet-induced Sp1 cholesterolgenic response, loss of cortical F-actin, and development of insulin resistance. Similarly, increases in HMGCR expression and cholesterol were measured in muscle from GFAT transgenic mice compared to age- and weight-match wildtype littermate control mice. In the GFAT Tg mice we found that these increases were alleviated by MTM. CONCLUSIONS These data identify increased HBP/Sp1 activity as an early mechanism of diet-induced insulin resistance. Therapies targeting this mechanism may decelerate T2D development.
Collapse
Affiliation(s)
- Jacob D Covert
- Department of Anatomy, Cell Biology and Physiology, Indianapolis, IN, United States; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Brian A Grice
- Department of Anatomy, Cell Biology and Physiology, Indianapolis, IN, United States; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Matthew G Thornburg
- Department of Anatomy, Cell Biology and Physiology, Indianapolis, IN, United States; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Manpreet Kaur
- Department of Anatomy, Cell Biology and Physiology, Indianapolis, IN, United States; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Andrew P Ryan
- Department of Anatomy, Cell Biology and Physiology, Indianapolis, IN, United States; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, United States; Eli Lilly and Company, Indianapolis, IN, United States
| | - Lixuan Tackett
- Department of Anatomy, Cell Biology and Physiology, Indianapolis, IN, United States; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Theja Bhamidipati
- Department of Anatomy, Cell Biology and Physiology, Indianapolis, IN, United States; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Natalie D Stull
- Indiana Biosciences Research Institute Indianapolis, IN, United States
| | - Teayoun Kim
- Comprehensive Diabetes Center and Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Kirk M Habegger
- Comprehensive Diabetes Center and Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Donald A McClain
- Section of Endocrinology and Metabolism, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Joseph T Brozinick
- Department of Anatomy, Cell Biology and Physiology, Indianapolis, IN, United States; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, United States; Comprehensive Diabetes Center and Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jeffrey S Elmendorf
- Department of Anatomy, Cell Biology and Physiology, Indianapolis, IN, United States; Department of Biochemistry and Molecular Biology, Indianapolis, IN, United States; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, United States.
| |
Collapse
|
19
|
Ding J, Lohman K, Molina A, Delbono O, Bertoni A, Shea S, Post W, Guo X, Barr RG, Manichaikul AW, Pankow JS, Rotter JI, Hoeschele I, Kritchevsky SB, Liu Y. The association between aging-related monocyte transcriptional networks and comorbidity burden: the Multi-Ethnic Study of Atherosclerosis (MESA). GeroScience 2023; 45:197-207. [PMID: 35737188 PMCID: PMC9886705 DOI: 10.1007/s11357-022-00608-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 06/16/2022] [Indexed: 02/03/2023] Open
Abstract
Translating our knowledge of the biological aging from animal models to humans may give rise to novel approaches of targeting multiple aging-related diseases simultaneously and increasing health span. Here, for the first time, we use transcriptomic signatures of monocytes to identify biological aging pathways underlying multiple aging-related diseases in humans. The ordinal logistic regression was used to cross-sectionally investigate transcriptomics of the comorbidity index in 1264 community-based Multi-Ethnic Study of Atherosclerosis (MESA) adults, 47% Caucasian, 32% Hispanic, 21% African American, and 51% female, aged 55-94 years. The comorbidity index was defined as the number of prevalent aging-related diseases including cardiovascular disease, type-2 diabetes, hypertension, cancer, dementia, chronic kidney disease, chronic obstructive pulmonary disease, and hip fracture. We identified 708 gene transcripts associated with the comorbidity index (FDR < 0.05) after adjusting for age, sex, ethnicity, and study site. In a weighted gene co-expression network analysis, as postulated, aging-related declines in apoptosis/autophagy (OR = 1.21 per SD increment, p = 0.0006) and ribosome/mitochondrion (OR = 0.90 per SD increment, p = 0.05) were positively associated with the comorbidity index. After adjusting for multiple comparisons, we identified 10 comorbidity-associated modules (FDR < 0.05), including the module of apoptosis/autophagy. There were three inter-correlated modules of these 10 involved in the complement subcomponent C1q, Fc gamma receptor I, and Fc gamma receptor III of the immune system, respectively. Aging-related upregulation of these three modules was positively associated with the comorbidity index. The odds of comorbidity increased with more of these modules acting together in a dose-response fashion. In conclusion, transcriptomic analysis of human immune cells may identify biomarker panels indicative of comprehensive biological mechanisms, especially immune signaling pathways, contributing to health aging.
Collapse
Affiliation(s)
- Jingzhong Ding
- Department of Internal Medicine, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC, 27157, USA.
| | - Kurt Lohman
- Department of Medicine, Duke University School of Medicine, Durham, NC, 27701, USA
| | - Anthony Molina
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Osvaldo Delbono
- Department of Internal Medicine, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC, 27157, USA
| | - Alain Bertoni
- Department of Epidemiology & Prevention, Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Steven Shea
- Departments of Medicine and Epidemiology, Columbia University, New York, NY, 10032, USA
| | - Wendy Post
- Departments of Pathology and Cardiology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - 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, 90502, USA
| | - R Graham Barr
- Department of Medicine, Columbia University, New York, NY, 10032, USA
| | - Ani W Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, 22908, USA
| | - James S Pankow
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, 55454, 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, 90502, USA
| | - Ina Hoeschele
- Biocomplexity Institute, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Stephen B Kritchevsky
- Department of Internal Medicine, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC, 27157, USA
| | - Yongmei Liu
- Department of Medicine, Duke University School of Medicine, Durham, NC, 27701, USA
| |
Collapse
|
20
|
Uramaru N, Kawashima A, Osabe M, Higuchi T. Rhododendrol, a reductive metabolite of raspberry ketone, suppresses the differentiation of 3T3‑L1 cells into adipocytes. Mol Med Rep 2023; 27:51. [PMID: 36633126 PMCID: PMC9879071 DOI: 10.3892/mmr.2023.12938] [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: 03/25/2022] [Accepted: 12/21/2022] [Indexed: 01/13/2023] Open
Abstract
Obesity is a serious medical condition worldwide, and a major risk factor for type 2 diabetes, metabolic syndrome, cancer and cardiovascular disease. In addition to changes in dietary habits and physical activity, consuming supplements to maintain good health and prevent obesity is important in modern society. Raspberry ketone (RK) is a natural phenolic ketone found in the European red raspberry (Rubus idaeus L.) and is hypothesized to prevent obesity when administered orally. The present study found that RK was reduced to rhododendrol (ROH) in human liver microsomes and cytosol. The present study investigated whether the metabolite ROH had anti‑adipogenic effects using mouse 3T3‑L1 cells. The effects of ROH or RK on lipid accumulation during differentiation of 3T3‑L1 pre‑adipocyte into adipocyte were determined using Oil Red O staining. CCAAT enhancer‑binding protein α (C/EBPα) and peroxisome proliferator‑activated receptor γ (PPARγ) mRNA and protein expression were examined using reverse transcription‑quantitative PCR and western blotting analysis, respectively. The present study revealed that ROH suppressed lipid accumulation in the cells, similar to RK. In addition, ROH suppressed the mRNA expression levels of C/EBPα and PPARγ in 3T3‑L1 adipocytes. Furthermore, ROH suppressed PPARγ protein expression in 3T3‑L1 adipocytes. These findings suggested that ROH is an active metabolite with an anti‑adipogenic effect, which may contribute to the anti‑obesity effect of orally administered RK. The present study indicated that it is important to understand the biological activity of the metabolites of orally administered compounds.
Collapse
Affiliation(s)
- Naoto Uramaru
- Division of Pharmaceutical Health Biosciences, Nihon Pharmaceutical University, Saitama 362-0806, Japan
| | - Azusa Kawashima
- Division of Pharmaceutical Health Biosciences, Nihon Pharmaceutical University, Saitama 362-0806, Japan
| | - Makoto Osabe
- Division of Pharmaceutical Health Biosciences, Nihon Pharmaceutical University, Saitama 362-0806, Japan
| | - Toshiyuki Higuchi
- Division of Pharmaceutical Health Biosciences, Nihon Pharmaceutical University, Saitama 362-0806, Japan,Correspondence to: Professor Toshiyuki Higuchi, Division of Pharmaceutical Health Biosciences, Nihon Pharmaceutical University, 10281 Komuro, Ina-machi, Kitaadachi-gun, Saitama 362-0806, Japan, E-mail:
| |
Collapse
|
21
|
Hong X, Wu Z, Cao W, Lv J, Yu C, Huang T, Sun D, Liao C, Pang Y, Pang Z, Cong L, Wang H, Wu X, Liu Y, Gao W, Li L. Longitudinal Association of DNA Methylation With Type 2 Diabetes and Glycemic Traits: A 5-Year Cross-Lagged Twin Study. Diabetes 2022; 71:2804-2817. [PMID: 36170668 DOI: 10.2337/db22-0513] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 09/20/2022] [Indexed: 01/11/2023]
Abstract
Investigators of previous cross-sectional epigenome-wide association studies (EWAS) in adults have reported hundreds of 5'-cytosine-phosphate-guanine-3' (CpG) sites associated with type 2 diabetes mellitus (T2DM) and glycemic traits. However, the results from EWAS have been inconsistent, and longitudinal observations of these associations are scarce. Furthermore, few studies have investigated whether DNA methylation (DNAm) could be modified by smoking, drinking, and glycemic traits, which have broad impacts on genome-wide DNAm and result in altering the risk of T2DM. Twin studies provide a valuable tool for epigenetic studies, as twins are naturally matched for genetic information. In this study, we conducted a systematic literature search in PubMed and Embase for EWAS, and 214, 33, and 117 candidate CpG sites were selected for T2DM, HbA1c, and fasting blood glucose (FBG). Based on 1,070 twins from the Chinese National Twin Registry, 67, 17, and 16 CpG sites from previous studies were validated for T2DM, HbA1c, and FBG. Longitudinal review and blood sampling for phenotypic information and DNAm were conducted twice in 2013 and 2018 for 308 twins. A cross-lagged analysis was performed to examine the temporal relationship between DNAm and T2DM or glycemic traits in the longitudinal data. A total of 11 significant paths from T2DM to subsequent DNAm and 15 paths from DNAm to subsequent T2DM were detected, suggesting both directions of associations. For glycemic traits, we detected 17 cross-lagged associations from baseline glycemic traits to subsequent DNAm, and none were from the other cross-lagged direction, indicating that CpG sites may be the consequences, not the causes, of glycemic traits. Finally, a longitudinal mediation analysis was performed to explore the mediation effects of DNAm on the associations of smoking, drinking, and glycemic traits with T2DM. No significant mediations of DNAm in the associations linking smoking and drinking with T2DM were found. In contrast, our study suggested a potential role of DNAm of cg19693031, cg00574958, and cg04816311 in mediating the effect of altered glycemic traits on T2DM.
Collapse
Affiliation(s)
- Xuanming Hong
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Zhiyu Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Weihua Cao
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Jun Lv
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Tao Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Dianjianyi Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Chunxiao Liao
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Yuanjie Pang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Zengchang Pang
- Qingdao Center for Disease Control and Prevention, Qingdao, China
| | - Liming Cong
- Zhejiang Center for Disease Control and Prevention, Hangzhou, China
| | - Hua Wang
- Jiangsu Center for Disease Control and Prevention, Nanjing, China
| | - Xianping Wu
- Sichuan Center for Disease Control and Prevention, Chengdu, China
| | - Yu Liu
- Heilongjiang Center for Disease Control and Prevention, Harbin, China
| | - Wenjing Gao
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| |
Collapse
|
22
|
Ye Z, Wang S, Huang X, Chen P, Deng L, Li S, Lin S, Wang Z, Liu B. Plasma Exosomal miRNAs Associated With Metabolism as Early Predictor of Gestational Diabetes Mellitus. Diabetes 2022; 71:2272-2283. [PMID: 35926094 PMCID: PMC9630082 DOI: 10.2337/db21-0909] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 08/02/2022] [Indexed: 01/25/2023]
Abstract
To date, the miRNA expression profile of plasma exosomes in women whose pregnancy is complicated by gestational diabetes mellitus (GDM) has not been fully clarified. In this study, differentially expressed miRNAs in plasma exosomes were identified by high-throughput small-RNA sequencing in 12 pregnant women with GDM and 12 with normal glucose tolerance (NGT) and validated in 102 pregnant women with GDM and 101 with NGT. A total of 22 exosomal miRNAs were found, five of which were verified by real-time qPCR. Exosomal miR-423-5p was upregulated, whereas miR-122-5p, miR-148a-3p, miR-192-5p, and miR-99a-5p were downregulated in women whose pregnancy was complicated by GDM. IGF1R and GYS1 as target genes of miR-423-5p, and G6PC3 and FDFT1 as target genes of miR-122-5p were associated with insulin and AMPK signaling pathways and may participate in the regulation of metabolism in GDM. The five exosomal miRNAs had an area under the curve of 0.82 (95%CI, 0.73, ∼0.91) in early prediction of GDM. Our study demonstrates that dysregulated exosomal miRNAs in plasma from pregnant women with GDM might influence the insulin and AMPK signaling pathways and could contribute to the early prediction of GDM.
Collapse
Affiliation(s)
- Zhixin Ye
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Songzi Wang
- Department of Laboratory Medicine, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Xiaoqing Huang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Peisong Chen
- Department of Laboratory Medicine, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Langhui Deng
- Department of Laboratory Medicine, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Shiqi Li
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Suiwen Lin
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Zilian Wang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Bin Liu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People’s Republic of China
- Corresponding author: Bin Liu,
| |
Collapse
|
23
|
Wu Z, Chen L, Hong X, Si J, Cao W, Yu C, Huang T, Sun D, Liao C, Pang Y, Pang Z, Cong L, Wang H, Wu X, Liu Y, Guo Y, Chen Z, Lv J, Gao W, Li L. Temporal associations between leukocytes DNA methylation and blood lipids: a longitudinal study. Clin Epigenetics 2022; 14:132. [PMID: 36274151 PMCID: PMC9588246 DOI: 10.1186/s13148-022-01356-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/13/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND The associations between blood lipids and DNA methylation have been investigated in epigenome-wide association studies mainly among European ancestry populations. Several studies have explored the direction of the association using cross-sectional data, while evidence of longitudinal data is still lacking. RESULTS We tested the associations between peripheral blood leukocytes DNA methylation and four lipid measures from Illumina 450 K or EPIC arrays in 1084 participants from the Chinese National Twin Registry and replicated the result in 988 participants from the China Kadoorie Biobank. A total of 23 associations of 19 CpG sites were identified, with 4 CpG sites located in or adjacent to 3 genes (TMEM49, SNX5/SNORD17 and CCDC7) being novel. Among the validated associations, we conducted a cross-lagged analysis to explore the temporal sequence and found temporal associations of methylation levels of 2 CpG sites with triglyceride and 2 CpG sites with high-density lipoprotein-cholesterol (HDL-C) in all twins. In addition, methylation levels of cg11024682 located in SREBF1 at baseline were temporally associated with triglyceride at follow-up in only monozygotic twins. We then performed a mediation analysis with the longitudinal data and the result showed that the association between body mass index and HDL-C was partially mediated by the methylation level of cg06500161 (ABCG1), with a mediation proportion of 10.1%. CONCLUSIONS Our study indicated that the DNA methylation levels of ABCG1, AKAP1 and SREBF1 may be involved in lipid metabolism and provided evidence for elucidating the regulatory mechanism of lipid homeostasis.
Collapse
Affiliation(s)
- Zhiyu Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
| | - Lu Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
| | - Xuanming Hong
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
| | - Jiahui Si
- National Institute of Health Data Science at Peking University, Peking University, Beijing, 100191, China
| | - Weihua Cao
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, 100191, China
| | - Tao Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
| | - Dianjianyi Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
| | - Chunxiao Liao
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
| | - Yuanjie Pang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
| | - Zengchang Pang
- Qingdao Center for Disease Control and Prevention, Qingdao, 266033, China
| | - Liming Cong
- Zhejiang Center for Disease Control and Prevention, Hangzhou, 310051, China
| | - Hua Wang
- Jiangsu Center for Disease Control and Prevention, Nanjing, 210008, China
| | - Xianping Wu
- Sichuan Center for Disease Control and Prevention, Chengdu, 610041, China
| | - Yu Liu
- Heilongjiang Center for Disease Control and Prevention, Harbin, 150090, China
| | - Yu Guo
- Fuwai hospital Chinese Academy of Medical Sciences, Beijing, 100037, China
| | - Zhengming Chen
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, OX3 7LF, UK
| | - Jun Lv
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100191, China
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, 100191, China
- Key Laboratory of Molecular Cardiovascular Sciences (Peking University), Ministry of Education, Beijing, 100191, China
| | - Wenjing Gao
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100191, China.
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100191, China.
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, 100191, China.
| |
Collapse
|
24
|
Parker DC, Wan M, Lohman K, Hou L, Nguyen AT, Ding J, Bertoni A, Shea S, Burke GL, Jacobs DR, Post W, Corcoran D, Hoeschele I, Parks JS, Liu Y. Monocyte miRNAs Are Associated With Type 2 Diabetes. Diabetes 2022; 71:853-861. [PMID: 35073575 PMCID: PMC8965663 DOI: 10.2337/db21-0704] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 01/20/2022] [Indexed: 11/13/2022]
Abstract
miRNAs are small noncoding RNAs that may contribute to common diseases through epigenetic regulation of gene expression. Little is known regarding the role of miRNAs in type 2 diabetes (T2D). We performed miRNA sequencing and transcriptomic profiling of peripheral monocytes from the longitudinal Multi-Ethnic Study of Atherosclerosis (MESA) (N = 1,154). We examined associations between miRNAs and prevalent impaired fasting glucose and T2D and evaluated the T2D-associated miRNA effect on incident T2D. Of 774 detected miRNAs, 6 (miR-22-3p, miR-33a-5p, miR-181c-5p, miR-92b-3p, miR-222-3p, and miR-944) were associated with prevalent T2D. For five of the six miRNAs (all but miR-222-3p), our findings suggest a dose-response relationship with impaired fasting glucose and T2D. Two of the six miRNAs were associated with incident T2D (miR-92b-3p: hazard ratio [HR] 1.64, P = 1.30E-03; miR-222-3p: HR 1.97, P = 9.10E-03) in the highest versus lowest tertile of expression. Most of the T2D-associated miRNAs were also associated with HDL cholesterol concentrations. The genes targeted by these miRNAs belong to key nodes of a cholesterol metabolism transcriptomic network. Higher levels of miRNA expression expected to increase intracellular cholesterol accumulation in monocytes are linked to an increase in T2D risk.
Collapse
Affiliation(s)
- Daniel C. Parker
- Division of Geriatrics, Department of Medicine, Duke University School of Medicine, Durham, NC
- Duke University Center for the Study of Aging and Human Development, Durham, NC
| | - Ma Wan
- Division of Cardiology, Department of Medicine, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC
| | - Kurt Lohman
- Division of Cardiology, Department of Medicine, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC
| | - Li Hou
- Division of Cardiology, Department of Medicine, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC
| | - Anh Tram Nguyen
- Division of Cardiology, Department of Medicine, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC
| | - Jingzhong Ding
- Wake Forest University School of Medicine, Winston-Salem, NC
| | - Alain Bertoni
- Wake Forest University School of Medicine, Winston-Salem, NC
| | - Steve Shea
- Columbia University School of Medicine, New York, NY
| | | | - David R. Jacobs
- University of Minnesota School of Public Health, Minneapolis, MN
| | - Wendy Post
- Johns Hopkins University School of Medicine, Baltimore, MD
| | - David Corcoran
- Duke Center for Genomic and Computational Biology, Duke University, Durham, NC
| | - Ina Hoeschele
- Department of Statistics and Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA
| | - John S. Parks
- Wake Forest University School of Medicine, Winston-Salem, NC
| | - Yongmei Liu
- Division of Cardiology, Department of Medicine, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC
| |
Collapse
|
25
|
Christiansen D, Bishop DJ. Aerobic-interval exercise with blood flow restriction potentiates early markers of metabolic health in man. Acta Physiol (Oxf) 2022; 234:e13769. [PMID: 34984835 DOI: 10.1111/apha.13769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/02/2021] [Accepted: 01/01/2022] [Indexed: 12/06/2022]
Abstract
AIM This study examined whether aerobic-interval exercise with blood flow restriction (BFR) potentiates early markers of metabolic health compared to exercise with systemic hypoxia or normoxia in man. METHODS In a randomized-crossover fashion, eight healthy men completed nine 2-minute running bouts at 105% of their lactate threshold on three occasions separated by one week, either with BFR (BFR-trial), systemic hypoxia (HYP-trial) or normoxia (control; CON-trial). Near-infrared spectroscopy was used to assess the muscle level of hypoxia. A muscle biopsy was collected at rest and 3 hours after exercise to quantify genes involved in cholesterol synthesis (PGC-1α2), glucose disposal (GLUT4) and capillary growth (HIF-1α; VEGFA), as well as mitochondrial respiration (PGC-1α2/3), uncoupling (UCP3) and expansion (p53; COXIV-1/2; CS; AMPKα1/2). RESULTS The muscle level of hypoxia was matched between the BFR-trial and HYP-trial (~90%; P > .05), which was greater than the CON-trial (~70%; P < .05). PGC-1α2 increased most in the BFR-trial (16-fold vs CON-trial; 11-fold vs HYP-trial; P < .05). GLUT4 and VEGFA selectively increased by 2.0 and 3.4-fold, respectively in BFR-trial (P < .05), which was greater than CON-trial (1.2 and 1.3 fold) and HYP-trial (1.2 and 1.8 fold; P < .05). UCP3 increased more in BFR-trial than the HYP-trial (4.3 vs 1.6 fold), but was not different between BFR-trial and CON-trial (2.1 fold) or between CON-trial and HYP-trial (P > .05). No trial differences were evident for other genes (P > .05). CONCLUSION Independent of the muscle level of hypoxia, BFR-exercise potentiates early markers of metabolic health associated with the regulation of cholesterol production and glucose homeostasis in man.
Collapse
Affiliation(s)
- Danny Christiansen
- Institute for Health & Sport Victoria University Melbourne Victoria Australia
| | - David J. Bishop
- Institute for Health & Sport Victoria University Melbourne Victoria Australia
| |
Collapse
|
26
|
Miroshnikova VV, Panteleeva AA, Pobozheva IA, Razgildina ND, Polyakova EA, Markov AV, Belyaeva OD, Berkovich OA, Baranova EI, Nazarenko MS, Puzyrev VP, Pchelina SN. ABCA1 and ABCG1 DNA methylation in epicardial adipose tissue of patients with coronary artery disease. BMC Cardiovasc Disord 2021; 21:566. [PMID: 34837967 PMCID: PMC8627066 DOI: 10.1186/s12872-021-02379-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 11/10/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Recent studies have focused on the potential role of epicardial adipose tissue (EAT) in the development of coronary artery disease (CAD). ABCA1 and ABCG1 transporters regulate cell cholesterol content and reverse cholesterol transport. We aimed to determine whether DNA methylation and mRNA levels of the ABCA1 and ABCG1 genes in EAT and subcutaneous adipose tissue (SAT) were associated with CAD. METHODS Paired EAT and SAT samples were collected from 82 patients undergoing elective cardiac surgery either for coronary artery bypass grafting (CAD group, N = 66) or valve surgery (NCAD group, N = 16). ABCA1 and ABCG1 mRNA levels in EAT and SAT samples were analyzed using real time polymerase chain reaction, ABCA1 protein levels in EAT samples were assessed by western blotting. ABCA1 and ABCG1 DNA methylation analysis was performed in 24 samples from the CAD group and 9 samples from the NCAD group via pyrosequencing. RESULTS DNA methylation levels in the ABCA1 promoter and ABCG1 cg27243685 and cg06500161 CpG sites were higher in EAT samples from patients with CAD compared with NCAD (21.92% vs 10.81%, p = 0.003; 71.51% vs 68.42%, p = 0.024; 46.11% vs 37.79%, p = 0.016, respectively). In patients with CAD, ABCA1 and ABCG1 DNA methylation levels were higher in EAT than in SAT samples (p < 0.05). ABCA1 mRNA levels in EAT samples were reduced in the subgroup of patients with CAD and concomitant carotid artery disease or peripheral artery disease compared with the NCAD group (p = 0.024). ABCA1 protein levels in EAT samples tended to be lower in CAD patients than in the NCAD group (p = 0.053). DNA methylation levels at the ABCG1 cg27243685 site positively correlated with plasma triglyceride concentration (r = 0.510, p = 0.008), body mass index (r = 0.556, p = 0.013) and waist-to-hip ratio (r = 0.504, p = 0.012) in SAT samples. CONCLUSION CAD is associated with ABCA1 and ABCG1 DNA hypermethylation in EAT. CAD with concomitant carotid artery disease or peripheral artery disease is accompanied by decreased ABCA1 gene expression in EAT. DNA methylation levels at the ABCG1 cg27243685 locus in SAT are associated with hypertriglyceridemia and obesity.
Collapse
Affiliation(s)
- Valentina V Miroshnikova
- Petersburg Nuclear Physics Institute Named By B.P. Konstantinov of National Research Center "Kurchatov Institute", Gatchina, Russian Federation.
- Pavlov First Saint Petersburg State Medical University, St.-Petersburg, Russian Federation.
| | - Alexandra A Panteleeva
- Petersburg Nuclear Physics Institute Named By B.P. Konstantinov of National Research Center "Kurchatov Institute", Gatchina, Russian Federation
- Pavlov First Saint Petersburg State Medical University, St.-Petersburg, Russian Federation
- National Research Centre "Kurchatov Institute", Moscow, Russia
| | - Irina A Pobozheva
- Petersburg Nuclear Physics Institute Named By B.P. Konstantinov of National Research Center "Kurchatov Institute", Gatchina, Russian Federation
- Pavlov First Saint Petersburg State Medical University, St.-Petersburg, Russian Federation
- National Research Centre "Kurchatov Institute", Moscow, Russia
| | - Natalia D Razgildina
- Petersburg Nuclear Physics Institute Named By B.P. Konstantinov of National Research Center "Kurchatov Institute", Gatchina, Russian Federation
| | - Ekaterina A Polyakova
- Pavlov First Saint Petersburg State Medical University, St.-Petersburg, Russian Federation
| | - Anton V Markov
- Laboratory of Population Genetics, Research Institute of Medical Genetics, Tomsk, Russian Federation
| | - Olga D Belyaeva
- Pavlov First Saint Petersburg State Medical University, St.-Petersburg, Russian Federation
| | - Olga A Berkovich
- Pavlov First Saint Petersburg State Medical University, St.-Petersburg, Russian Federation
| | - Elena I Baranova
- Pavlov First Saint Petersburg State Medical University, St.-Petersburg, Russian Federation
| | - Maria S Nazarenko
- Laboratory of Population Genetics, Research Institute of Medical Genetics, Tomsk, Russian Federation
| | - Valery P Puzyrev
- Laboratory of Population Genetics, Research Institute of Medical Genetics, Tomsk, Russian Federation
| | - Sofya N Pchelina
- Petersburg Nuclear Physics Institute Named By B.P. Konstantinov of National Research Center "Kurchatov Institute", Gatchina, Russian Federation
- Pavlov First Saint Petersburg State Medical University, St.-Petersburg, Russian Federation
- National Research Centre "Kurchatov Institute", Moscow, Russia
| |
Collapse
|
27
|
Hulme KD, Noye EC, Short KR, Labzin LI. Dysregulated Inflammation During Obesity: Driving Disease Severity in Influenza Virus and SARS-CoV-2 Infections. Front Immunol 2021; 12:770066. [PMID: 34777390 PMCID: PMC8581451 DOI: 10.3389/fimmu.2021.770066] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 09/30/2021] [Indexed: 12/15/2022] Open
Abstract
Acute inflammation is a critical host defense response during viral infection. When dysregulated, inflammation drives immunopathology and tissue damage. Excessive, damaging inflammation is a hallmark of both pandemic influenza A virus (IAV) infections and Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2) infections. Chronic, low-grade inflammation is also a feature of obesity. In recent years, obesity has been recognized as a growing pandemic with significant mortality and associated costs. Obesity is also an independent risk factor for increased disease severity and death during both IAV and SARS-CoV-2 infection. This review focuses on the effect of obesity on the inflammatory response in the context of viral respiratory infections and how this leads to increased viral pathology. Here, we will review the fundamentals of inflammation, how it is initiated in IAV and SARS-CoV-2 infection and its link to disease severity. We will examine how obesity drives chronic inflammation and trained immunity and how these impact the immune response to IAV and SARS-CoV-2. Finally, we review both medical and non-medical interventions for obesity, how they impact on the inflammatory response and how they could be used to prevent disease severity in obese patients. As projections of global obesity numbers show no sign of slowing down, future pandemic preparedness will require us to consider the metabolic health of the population. Furthermore, if weight-loss alone is insufficient to reduce the risk of increased respiratory virus-related mortality, closer attention must be paid to a patient’s history of health, and new therapeutic options identified.
Collapse
Affiliation(s)
- Katina D Hulme
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Ellesandra C Noye
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Kirsty R Short
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Larisa I Labzin
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia.,Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
28
|
Comparison and Analysis on the Existing Single-Herbal Strategies against Viral Myocarditis. Genet Res (Camb) 2021; 2021:9952620. [PMID: 34456633 PMCID: PMC8371739 DOI: 10.1155/2021/9952620] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 07/31/2021] [Indexed: 02/08/2023] Open
Abstract
Purpose Herbal medicine is one of crucial symbols of Chinese national medicine. Investigation on molecular responses of different herbal strategies against viral myocarditis is immeasurably conducive to targeting drug development in the current international absence of miracle treatment. Methods Literature retrieval platforms were applied in the collection of existing empirical evidences for viral myocarditis-related single-herbal strategies. SwissTargetPrediction, Metascape, and Discovery Studio coordinating with multidatabases investigated underlying target genes, interactive proteins, and docking molecules in turn. Results Six single-herbal medicines consisting of Huangqi (Hedysarum Multijugum Maxim), Yuganzi (Phyllanthi Fructus), Kushen (Sophorae Flavescentis Radix), Jianghuang (Curcumaelongae Rhizoma), Chaihu (Radix Bupleuri), and Jixueteng (Spatholobus Suberectus Dunn) meet the requirement. There were 11 overlapped and 73 unique natural components detected in these herbs. SLC6A2, SLC6A4, NOS2, PPARA, PPARG, ACHE, CYP2C19, CYP51A1, and CHRM2 were equally targeted by six herbs and identified as viral myocarditis-associated symbols. MCODE algorithm exposed the hub role of SRC and EGFR in strategies without Jianghuang. Subsequently, we learned intermolecular interactions of herbal components and their targeting heart-tissue-specific CHRM2, FABP3, TNNC1, TNNI3, TNNT2, and SCN5A and cardiac-myocytes-specific IL6, MMP1, and PLAT coupled with viral myocarditis. Ten interactive characteristics such as π-alkyl and van der Waals were modeled in which ARG111, LYS253, ILE114, and VAL11 on cardiac troponin (TNNC1-TNNI3-TNNT2) and ARG208, ASN106, and ALA258 on MMP1 fulfilled potential communicating anchor with ellagic acid, 5α, 9α-dihydroxymatrine, and leachianone g via hydrogen bond and hydrophobic interaction, respectively. Conclusions The comprehensive outcomes uncover differences and linkages between six herbs against viral myocarditis through component and target analysis, fostering development of drugs.
Collapse
|
29
|
Bries AE, Webb JL, Vogel B, Carrillo C, Day TA, Kimber MJ, Valentine RJ, Rowling MJ, Clark S, Schalinske KL, McNeill EM. RNA Sequencing Reveals Key Metabolic Pathways Are Modified by Short-Term Whole Egg Consumption. Front Nutr 2021; 8:652192. [PMID: 34041258 PMCID: PMC8141817 DOI: 10.3389/fnut.2021.652192] [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: 01/11/2021] [Accepted: 04/06/2021] [Indexed: 11/13/2022] Open
Abstract
Eggs are protein-rich, nutrient-dense, and contain bioactive ingredients that have been shown to modify gene expression and impact health. To understand the effects of egg consumption on tissue-specific mRNA and microRNA expression, we examined the role of whole egg consumption (20% protein, w/w) on differentially expressed genes (DEGs) between rat (n = 12) transcriptomes in the prefrontal cortex (PFC), liver, kidney, and visceral adipose tissue (VAT). Principal component analysis with hierarchical clustering was used to examine transcriptome profiles between dietary treatment groups. We performed Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis as well as genetic network and disease enrichment analysis to examine which metabolic pathways were the most predominantly altered in each tissue. Overall, our data demonstrates that whole egg consumption for 2 weeks modified the expression of 52 genes in the PFC, 22 genes in VAT, and two genes in the liver (adj p < 0.05). Additionally, 16 miRNAs were found to be differentially regulated in the PFC, VAT, and liver, but none survived multiple testing correction. The main pathways influenced by WE consumption were glutathione metabolism in VAT and cholesterol biosynthesis in the PFC. These data highlight key pathways that may be involved in diseases and are impacted by acute consumption of a diet containing whole eggs.
Collapse
Affiliation(s)
- Amanda E. Bries
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, United States
- Interdepartmental Graduate Program in Nutritional Sciences, Iowa State University, Ames, IA, United States
| | - Joe L. Webb
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, United States
- Interdepartmental Graduate Program in Nutritional Sciences, Iowa State University, Ames, IA, United States
| | - Brooke Vogel
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, United States
| | - Claudia Carrillo
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, United States
| | - Timothy A. Day
- Department of Biomedical Sciences, Iowa State University College of Veterinary Medicine, Ames, IA, United States
| | - Michael J. Kimber
- Department of Biomedical Sciences, Iowa State University College of Veterinary Medicine, Ames, IA, United States
| | - Rudy J. Valentine
- Interdepartmental Graduate Program in Nutritional Sciences, Iowa State University, Ames, IA, United States
- Department of Kinesiology, Iowa State University, Ames, IA, United States
| | - Matthew J. Rowling
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, United States
- Interdepartmental Graduate Program in Nutritional Sciences, Iowa State University, Ames, IA, United States
| | - Stephanie Clark
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, United States
| | - Kevin L. Schalinske
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, United States
- Interdepartmental Graduate Program in Nutritional Sciences, Iowa State University, Ames, IA, United States
| | - Elizabeth M. McNeill
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, United States
- Interdepartmental Graduate Program in Nutritional Sciences, Iowa State University, Ames, IA, United States
| |
Collapse
|
30
|
Fischer MA, Vondriska TM. Clinical epigenomics for cardiovascular disease: Diagnostics and therapies. J Mol Cell Cardiol 2021; 154:97-105. [PMID: 33561434 PMCID: PMC8330446 DOI: 10.1016/j.yjmcc.2021.01.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 01/05/2021] [Accepted: 01/10/2021] [Indexed: 12/28/2022]
Abstract
The study of epigenomics has advanced in recent years to span the regulation of a single genetic locus to the structure and orientation of entire chromosomes within the nucleus. In this review, we focus on the challenges and opportunities of clinical epigenomics in cardiovascular disease. As an integrator of genetic and environmental inputs, and because of advances in measurement techniques that are highly reproducible and provide sequence information, the epigenome is a rich source of potential biosignatures of cardiovascular health and disease. Most of the studies to date have focused on the latter, and herein we discuss observations on epigenomic changes in human cardiovascular disease, examining the role of protein modifiers of chromatin, noncoding RNAs and DNA modification. We provide an overview of cardiovascular epigenomics, discussing the challenges of data sovereignty, data analysis, doctor-patient ethics and innovations necessary to implement precision health.
Collapse
Affiliation(s)
- Matthew A Fischer
- Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine at UCLA, USA.
| | - Thomas M Vondriska
- Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine at UCLA, USA
| |
Collapse
|
31
|
Cohain AT, Barrington WT, Jordan DM, Beckmann ND, Argmann CA, Houten SM, Charney AW, Ermel R, Sukhavasi K, Franzen O, Koplev S, Whatling C, Belbin GM, Yang J, Hao K, Kenny EE, Tu Z, Zhu J, Gan LM, Do R, Giannarelli C, Kovacic JC, Ruusalepp A, Lusis AJ, Bjorkegren JLM, Schadt EE. An integrative multiomic network model links lipid metabolism to glucose regulation in coronary artery disease. Nat Commun 2021; 12:547. [PMID: 33483510 PMCID: PMC7822923 DOI: 10.1038/s41467-020-20750-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 12/08/2020] [Indexed: 01/30/2023] Open
Abstract
Elevated plasma cholesterol and type 2 diabetes (T2D) are associated with coronary artery disease (CAD). Individuals treated with cholesterol-lowering statins have increased T2D risk, while individuals with hypercholesterolemia have reduced T2D risk. We explore the relationship between lipid and glucose control by constructing network models from the STARNET study with sequencing data from seven cardiometabolic tissues obtained from CAD patients during coronary artery by-pass grafting surgery. By integrating gene expression, genotype, metabolomic, and clinical data, we identify a glucose and lipid determining (GLD) regulatory network showing inverse relationships with lipid and glucose traits. Master regulators of the GLD network also impact lipid and glucose levels in inverse directions. Experimental inhibition of one of the GLD network master regulators, lanosterol synthase (LSS), in mice confirms the inverse relationships to glucose and lipid levels as predicted by our model and provides mechanistic insights.
Collapse
Affiliation(s)
- Ariella T Cohain
- Department of Genetics and Genomic Science and Institute for Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - William T Barrington
- Department of Human Genetics/Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Daniel M Jordan
- Department of Genetics and Genomic Science and Institute for Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Noam D Beckmann
- Department of Genetics and Genomic Science and Institute for Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Carmen A Argmann
- Department of Genetics and Genomic Science and Institute for Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sander M Houten
- Department of Genetics and Genomic Science and Institute for Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Alexander W Charney
- Department of Genetics and Genomic Science and Institute for Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Raili Ermel
- Department of Cardiac Surgery, Tartu University Hospital, Tartu, Estonia
| | | | - Oscar Franzen
- Integrated Cardio Metabolic Centre, Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden
| | - Simon Koplev
- Department of Genetics and Genomic Science and Institute for Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Carl Whatling
- Translational Science, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Gillian M Belbin
- Department of Genetics and Genomic Science and Institute for Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jialiang Yang
- Department of Genetics and Genomic Science and Institute for Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ke Hao
- Department of Genetics and Genomic Science and Institute for Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Eimear E Kenny
- Department of Genetics and Genomic Science and Institute for Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Zhidong Tu
- Department of Genetics and Genomic Science and Institute for Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jun Zhu
- Department of Genetics and Genomic Science and Institute for Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Li-Ming Gan
- Early Clinical Development, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Ron Do
- Department of Genetics and Genomic Science and Institute for Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Chiara Giannarelli
- Department of Genetics and Genomic Science and Institute for Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Cardiovascular Research Centre, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jason C Kovacic
- Cardiovascular Research Centre, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Arno Ruusalepp
- Department of Cardiac Surgery, Tartu University Hospital, Tartu, Estonia
| | - Aldons J Lusis
- Department of Human Genetics/Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Johan L M Bjorkegren
- Department of Genetics and Genomic Science and Institute for Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Clinical Gene Networks AB, Stockholm, Sweden.
| | - Eric E Schadt
- Department of Genetics and Genomic Science and Institute for Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Sema4, Stamford, CT, USA.
| |
Collapse
|
32
|
Justice AE, Chittoor G, Gondalia R, Melton PE, Lim E, Grove ML, Whitsel EA, Liu CT, Cupples LA, Fernandez-Rhodes L, Guan W, Bressler J, Fornage M, Boerwinkle E, Li Y, Demerath E, Heard-Costa N, Levy D, Stewart JD, Baccarelli A, Hou L, Conneely K, Mori TA, Beilin LJ, Huang RC, Gordon-Larsen P, Howard AG, North KE. Methylome-wide association study of central adiposity implicates genes involved in immune and endocrine systems. Epigenomics 2020; 12:1483-1499. [PMID: 32901515 PMCID: PMC7923253 DOI: 10.2217/epi-2019-0276] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 05/22/2020] [Indexed: 12/14/2022] Open
Abstract
Aim: We conducted a methylome-wide association study to examine associations between DNA methylation in whole blood and central adiposity and body fat distribution, measured as waist circumference, waist-to-hip ratio and waist-to-height ratio adjusted for body mass index, in 2684 African-American adults in the Atherosclerosis Risk in Communities study. Materials & methods: We validated significantly associated cytosine-phosphate-guanine methylation sites (CpGs) among adults using the Women's Health Initiative and Framingham Heart Study participants (combined n = 5743) and generalized associations in adolescents from The Raine Study (n = 820). Results & conclusion: We identified 11 CpGs that were robustly associated with one or more central adiposity trait in adults and two in adolescents, including CpG site associations near TXNIP, ADCY7, SREBF1 and RAP1GAP2 that had not previously been associated with obesity-related traits.
Collapse
Affiliation(s)
- Anne E Justice
- Department of Population Health Sciences, Geisinger, Danville, PA 17822, USA
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Geetha Chittoor
- Department of Population Health Sciences, Geisinger, Danville, PA 17822, USA
| | - Rahul Gondalia
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Phillip E Melton
- School of Biomedical Science, Faculty of Health & Medical Sciences, The University of Western Australia, Perth, WA 6000, Australia
- School of Pharmacy & Biomedical Sciences, Faculty of Health Sciences, Curtin University, MRF Building, Perth, WA 6000, Australia
- Menzies Institute for Medical Research, College of Health & Medicine, University of Tasmania, Hobart, TA, 7000 Australia
| | - Elise Lim
- Department of Biostatistics, Boston University, Boston, MA 02118, USA
| | - Megan L Grove
- Human Genetics Center, Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Eric A Whitsel
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University, Boston, MA 02118, USA
| | - L Adrienne Cupples
- Department of Biostatistics, Boston University, Boston, MA 02118, USA
- Framingham Heart Study, Framingham, MA, 01701, USA
| | - Lindsay Fernandez-Rhodes
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Biobehavioral Health, Pennsylvania State University, University Park, PA 16802, USA
| | - Weihua Guan
- Division of Biostatistics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jan Bressler
- Human Genetics Center, Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Myriam Fornage
- Center for Human Genetics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030, USA
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yun Li
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Ellen Demerath
- Division of Epidemiology & Community Health, University of Minnesota, Minneapolis, MN 55455, USA
| | - Nancy Heard-Costa
- Framingham Heart Study, Framingham, MA, 01701, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Dan Levy
- Population sciences branch, NHLBI Framingham Heart Study, Framingham, MA 01702, USA
- Department of Medicine, Boston University, Boston, MA 02118, USA
| | - James D Stewart
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Andrea Baccarelli
- Laboratory of Environmental Epigenetics, Departments of Environmental Health Sciences & Epidemiology, Columbia University Mailman School of Public Health, New York, NY 10032, USA
| | - Lifang Hou
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University Chicago, Evanston, IL, USA
| | - Karen Conneely
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Trevor A Mori
- Medical School, University of Western Australia, Perth, Australia
| | | | - Rae-Chi Huang
- Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - Penny Gordon-Larsen
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, NC 27599, USA
- Carolina Population Center, University of North Carolina at Chapel Hill, NC 27516, USA
| | - Annie Green Howard
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Carolina Population Center, University of North Carolina at Chapel Hill, NC 27516, USA
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Carolina Population Center, University of North Carolina at Chapel Hill, NC 27516, USA
| |
Collapse
|
33
|
Dong X, Zhu Y, Wang S, Luo Y, Lu S, Nan F, Sun G, Sun X. Bavachinin inhibits cholesterol synthesis enzyme FDFT1 expression via AKT/mTOR/SREBP-2 pathway. Int Immunopharmacol 2020; 88:106865. [PMID: 32827918 DOI: 10.1016/j.intimp.2020.106865] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 07/20/2020] [Accepted: 07/29/2020] [Indexed: 12/15/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a progressive and chronic liver disease. No effective drug is currently approved for the treatment of NAFLD. Traditionally it is thought that pathogenesis of NAFLD develops from some imbalance in lipid control, thereby leading to hepatotoxicity and disease development. Squalene synthase (SQS), encoded by FDFT1, is a key regulator in cholesterol synthesis and thus a potential target for the treatment of NAFLD. Here we could identify bavachinin, a component from traditional Chinese medicine Fructus Psoraleae (FP), which apparently protects HepaRG cells from palmitic acid induced death, suppressing lipid accumulation and cholesterol synthesis through inhibition of FDFT1 through the AKT/mTOR/SREBP-2 pathway. Over-expression of FDFT1 abolished bavachinin (BVC) -induced inhibition of cholesterol synthesis. The data presented here suggest that bavachinin acts as a cholesterol synthesis enzyme inhibitor, and might serve as a drug for treating NAFLD in the future.
Collapse
Affiliation(s)
- Xi Dong
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, PR China; Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, PR China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China; Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, PR China
| | - Yue Zhu
- Harbin University of Commerce, Harbin, PR China
| | - Shan Wang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, PR China; Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, PR China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China; Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, PR China
| | - Yun Luo
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, PR China; Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, PR China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China; Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, PR China
| | - Shan Lu
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, PR China; Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, PR China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China; Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, PR China
| | - Fengwei Nan
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, PR China
| | - Guibo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, PR China; Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, PR China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China; Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, PR China.
| | - Xiaobo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, PR China; Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, PR China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China; Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, PR China.
| |
Collapse
|
34
|
The transcriptome of CMML monocytes is highly inflammatory and reflects leukemia-specific and age-related alterations. Blood Adv 2020; 3:2949-2961. [PMID: 31648319 DOI: 10.1182/bloodadvances.2019000585] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 08/23/2019] [Indexed: 12/19/2022] Open
Abstract
Chronic myelomonocytic leukemia (CMML) is an aggressive myeloid neoplasm of older individuals characterized by persistent monocytosis. Somatic mutations in CMML are heterogeneous and only partially explain the variability in clinical outcomes. Recent data suggest that cardiovascular morbidity is increased in CMML and contributes to reduced survival. Clonal hematopoiesis of indeterminate potential (CHIP), the presence of mutated blood cells in hematologically normal individuals, is a precursor of age-related myeloid neoplasms and associated with increased cardiovascular risk. To isolate CMML-specific alterations from those related to aging, we performed RNA sequencing and DNA methylation profiling on purified monocytes from CMML patients and from age-matched (old) and young healthy controls. We found that the transcriptional signature of CMML monocytes is highly proinflammatory, with upregulation of multiple inflammatory pathways, including tumor necrosis factor and interleukin (IL)-6 and -17 signaling, whereas age per se does not significantly contribute to this pattern. We observed no consistent correlations between aberrant gene expression and CpG island methylation, suggesting that proinflammatory signaling in CMML monocytes is governed by multiple and complex regulatory mechanisms. We propose that proinflammatory monocytes contribute to cardiovascular morbidity in CMML patients and promote progression by selection of mutated cell clones. Our data raise questions of whether asymptomatic patients with CMML benefit from monocyte-depleting or anti-inflammatory therapies.
Collapse
|
35
|
Zhang R, Wang J, Liu B, Wang W, Fan X, Zheng B, Yuan Q, Xue M, Xu F, Guo P, Chen Y. Differentially expressed lncRNAs, miRNAs and mRNAs with associated ceRNA networks in a mouse model of myocardial ischemia/reperfusion injury. Mol Med Rep 2020; 22:2487-2495. [PMID: 32705277 PMCID: PMC7411395 DOI: 10.3892/mmr.2020.11300] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 06/05/2020] [Indexed: 12/17/2022] Open
Abstract
Non‑coding RNAs, including long non‑coding RNAs (lncRNAs) and microRNAs (miRNAs/miRs), have significant regulatory effects on a number of biological processes in myocardial ischemia/reperfusion (I/R) injury, including cell differentiation, proliferation and apoptosis. In the present study, the expression levels of lncRNAs, miRNAs and mRNAs were evaluated in a mouse model of myocardial I/R injury. The potential functions of these differentially expressed genes were then analyzed via Gene Ontology and pathway analyses. Additionally, the interactions between lncRNA‑miRNA‑mRNA were predicted by constructing a competing endogenous RNA regulatory network. It was found that 14,366 lncRNAs, 151 miRNAs and 9,377 mRNAs were differentially expressed in mice hearts after I/R compared with the Sham group (fold change >2; P<0.05). The results indicated that these differentially expressed genes were involved in multiple molecular functions, including 'guanosine diphosphate binding', 'RNA polymerase II carboxy‑terminal domain kinase activity', 'TATA‑binding protein‑class protein binding', 'nicotinamide adenine dinucleotide binding' and 'protein phosphatase type 2A regulator activity'. The interactions between lncRNA‑miRNA‑mRNA, including five lncRNAs, 38 miRNAs and 196 mRNAs, were predicted, specifically Gm12040‑mmu‑miR‑125a‑5p‑decapping mRNA 1B, Rpl7l1‑ps1‑mmu‑miR‑124‑3p‑G protein‑coupled receptor 146, Gm11407‑mmu‑miR‑190a‑5p‑homeobox and leucine zipper encoding (HOMEZ), 1600029O15Rik‑mmu‑miR‑132‑3p‑HOMEZ and AK155692‑mmu‑miR‑1224‑3p‑activating transcription factor 6β. Collectively, these findings provided novel insights for future research on lncRNAs, miRNAs and mRNAs in myocardial I/R injury.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Emergency Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Jiali Wang
- Department of Emergency Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Baoshan Liu
- Department of Emergency Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Wenjun Wang
- Department of Emergency Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xinhui Fan
- Department of Emergency Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Boyuan Zheng
- Department of Emergency Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Qiuhuan Yuan
- Department of Emergency Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Mengyang Xue
- Department of Emergency Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Feng Xu
- Department of Emergency Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Ping Guo
- Department of Emergency Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yuguo Chen
- Department of Emergency Medicine, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
36
|
Chua NK, Coates HW, Brown AJ. Squalene monooxygenase: a journey to the heart of cholesterol synthesis. Prog Lipid Res 2020; 79:101033. [DOI: 10.1016/j.plipres.2020.101033] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/21/2020] [Accepted: 04/24/2020] [Indexed: 02/07/2023]
|
37
|
Badi SA, Motahhary A, Bahramali G, Masoumi M, Khalili SFS, Ebrahimzadeh N, Nouri P, Rahimi A, Masotti A, Moshiri A, Siadat SD. The regulation of Niemann-Pick C1-Like 1 (NPC1L1) gene expression in opposite direction by Bacteroides spp. and related outer membrane vesicles in Caco-2 cell line. J Diabetes Metab Disord 2020; 19:415-422. [PMID: 32550192 DOI: 10.1007/s40200-020-00522-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 03/24/2020] [Indexed: 11/28/2022]
Abstract
Purpose The intestine has substantial role in cholesterol homeostasis due to the presence of various cholesterol transporters and gut microbiota. Bacteroides spp. are important members of gut microbiota that employ outer membrane vesicles (OMVs) to interact with host. In this regard, we evaluated the effect of Bacteroides fragilis, Bacteroides thetaiotaomicron and related OMVs on the gene expression of important cholesterol transporters, Niemann-Pick C1-Like 1 (NPC1L1), ATP-binding cassette (ABCA1), and liver X receptors (LXRs) in Caco-2 cells. Methods OMVs were isolated from overnight brain heart infusion (BHI) broth of bacterial standard strains using deoxycholate and assessed by Scanning electron microscopy (SEM). The relative change in genes expression was assessed by Quantitative reverse transcription PCR (RT-qPCR) based on SYBR Green and 2-∆∆ct method in Caco-2 cells that were treated with bacteria and OMVs. Data were statistically analyzed with GraphPad Prism software. Finally, pathway enrichment based on the studied genes was performed using Cytoscape plugin ClueGO. Results B. fragilis (P value = 0.002) and B. thetaiotaomicron (P value = 0.001) significantly reduced NPC1L1 gene expression in Caco-2 cells. Interestingly, NPC1L1 transcripts were significantly increased by both OMVs(P value = 0.04) (P value = 0.01). Also, LXRβ was significantly down regulated by B. thetaiotaomicron (P value = 0.02). ClueGO analysis on the studied genes demonstrated several functional groups which involve in lipid and cholesterol metabolism. Conclusion The opposite effect of B. fragilis, B. thetaiotaomicron and related OMVs on the NPC1L1 gene expression was observed in Caco-2 cells. Interestingly, these effects partially were in line with the alternation of LXRs expression. However, based on pathway enrichment analysis, further molecular investigations are required to elaborate in details the specific association between Bacteroides spp. and OMVs with regulation of cholesterol signaling pathways including cholesterol transport, lipid storage, lipid homeostasis and cholesterol homeostasis.
Collapse
Affiliation(s)
- Sara Ahmadi Badi
- Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Atiyyeh Motahhary
- Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Golnaz Bahramali
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Morteza Masoumi
- Mycobacteriology and Pulmonary Research Department, Pasteur Institute of Iran, Tehran, Iran
| | | | - Nayereh Ebrahimzadeh
- Mycobacteriology and Pulmonary Research Department, Pasteur Institute of Iran, Tehran, Iran
| | - Pegah Nouri
- Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Ayoub Rahimi
- Mycobacteriology and Pulmonary Research Department, Pasteur Institute of Iran, Tehran, Iran
| | - Andrea Masotti
- Bambino Gesù Children's Hospital-IRCCS, Research Laboratories, Rome, Italy
| | - Arfa Moshiri
- Cancer Department, Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Seyed Davar Siadat
- Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran.,Mycobacteriology and Pulmonary Research Department, Pasteur Institute of Iran, Tehran, Iran.,Endocrinologyand Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
38
|
Bioinformatical Analysis of miRNA-mRNA Interaction Network Underlying Macrophage Aging and Cholesterol-Responsive Difference between Young and Aged Macrophages. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9267475. [PMID: 32626771 PMCID: PMC7306864 DOI: 10.1155/2020/9267475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/28/2020] [Accepted: 05/18/2020] [Indexed: 01/28/2023]
Abstract
Purpose Macrophage aging is involved with the occurrence and progression of age-related macular degeneration (AMD). The purpose of this study was to identify the specific microRNAs (miRNA), mRNAs, and their interactions underlying macrophage aging and response to cholesterol through bioinformatical analysis in order to get a better understanding of the mechanism of AMD. Methods The microarray data were obtained from Gene Expression Omnibus (accession GSE111304 and GSE111382). The age-related differentially expressed genes in macrophages were identified using R software. Further miRNA-mRNA interactions were analyzed through miRWalk, mirTarBase, starBase, and then produced by Cytoscape. The functional annotations including Gene Ontology and KEGG pathways of the miRNA target genes were performed by the DAVID and the STRING database. In addition, protein-protein interaction network was constructed to identify the key genes in response to exogenous cholesterol. Results When comparing aged and young macrophages, a total of 14 miRNAs and 101 mRNAs were detected as differentially expressed. Besides, 19 validated and 544 predicted miRNA-mRNA interactions were detected. Lipid metabolic process was found to be associated with macrophage aging through functional annotations of the miRNA targets. After being treated with oxidized and acetylated low-density lipoprotein, miR-714 and 16 mRNAs differentially expressed in response to both kinds of cholesterol between aged and young macrophages. Among them, 6 miRNA-mRNA predicted pairs were detected. The functional annotations were mainly related to lipid metabolism process and farnesyl diphosphate farnesyl transferase 1 (FDFT1) was identified to be the key gene in the difference of response to cholesterol between aged and young macrophages. Conclusions Lipid metabolic process was critical in both macrophage aging and response to cholesterol thus was regarded to be associated with the occurrence and progression of AMD. Moreover, miR-714-FDFT1 may modulate cholesterol homeostasis in aged macrophages and have the potential to be a novel therapeutic target for AMD.
Collapse
|
39
|
Bekkering S, Saner C, Riksen NP, Netea MG, Sabin MA, Saffery R, Stienstra R, Burgner DP. Trained Immunity: Linking Obesity and Cardiovascular Disease across the Life-Course? Trends Endocrinol Metab 2020; 31:378-389. [PMID: 32305098 DOI: 10.1016/j.tem.2020.01.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/17/2019] [Accepted: 01/09/2020] [Indexed: 02/06/2023]
Abstract
Obesity, a chronic inflammatory disease, is the most prevalent modifiable risk factor for cardiovascular disease. The mechanisms underlying inflammation in obesity are incompletely understood. Recent developments have challenged the dogma of immunological memory occurring exclusively in the adaptive immune system and show that the innate immune system has potential to be reprogrammed. This innate immune memory (trained immunity) is characterized by epigenetic and metabolic reprogramming of myeloid cells following endogenous or exogenous stimulation, resulting in enhanced inflammation to subsequent stimuli. Trained immunity phenotypes have now been reported for other immune and non-immune cells. Here, we provide a novel perspective on the putative role of trained immunity in mediating the adverse cardiovascular effects of obesity and highlight potential translational pathways.
Collapse
Affiliation(s)
- Siroon Bekkering
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia; Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Christoph Saner
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia; Department of Endocrinology, The Royal Children's Hospital, Parkville, Victoria, Australia
| | - Niels P Riksen
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands; Department for Immunology & Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Matthew A Sabin
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia; Department of Endocrinology, The Royal Children's Hospital, Parkville, Victoria, Australia; Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Richard Saffery
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia; Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Rinke Stienstra
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands; Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - David P Burgner
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia; Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
40
|
Zou T, Wang B, Li S, Liu Y, You J. Dietary apple polyphenols promote fat browning in high-fat diet-induced obese mice through activation of adenosine monophosphate-activated protein kinase α. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2020; 100:2389-2398. [PMID: 31916584 DOI: 10.1002/jsfa.10248] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 12/22/2019] [Accepted: 12/24/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Promoting brown and beige adipogenesis contributes to adaptive thermogenesis, which provides a defense against obesity and related disorders. Apple polyphenols (APs) play a significant role in treating variety of metabolic diseases. This study was conducted to determine the effects of APs on the development of brown and beige adipocytes and thermogenesis and investigate whether these effects are mediated by adenosine monophosphate-activated protein kinase (AMPK). High-fat diet (HFD)-induced obese mice and differentiated 3T3-L1 adipocytes were subjected to APs treatment. The thermogenic program and associated regulatory factors, and the involvement of AMPKα was assessed. RESULTS Dietary APs supplementation reduced adiposity and improved insulin sensitivity in HFD-induced obese mice. Moreover, APs increased the oxygen consumption and heat production and decreased respiratory exchange ratio, which were accompanied by the upregulation of thermogenic genes expression and the activation of AMPKα in brown fat and inguinal white fat. Further, APs treatment directly increased expression of brown adipogenic markers and induced phosphorylation of AMPKα in differentiated 3T3-L1 adipocytes, whereas the beneficial effects of APs were reversed by AMPK inhibition. CONCLUSION Our results provide new insights into the function of APs in regulating brown/beige adipogenesis and adaptive thermogenesis and suggest the potential application of APs in the prevention and therapeutics of obesity and associated metabolic diseases. © 2020 Society of Chemical Industry.
Collapse
Affiliation(s)
- Tiande Zou
- Jiangxi Province Key Laboratory of Animal Nutrition, Engineering Research Center of Feed Development, Jiangxi Agricultural University, Nanchang, China
| | - Bo Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shuo Li
- Jiangxi Province Key Laboratory of Animal Nutrition, Engineering Research Center of Feed Development, Jiangxi Agricultural University, Nanchang, China
| | - Yue Liu
- Jiangxi Province Key Laboratory of Animal Nutrition, Engineering Research Center of Feed Development, Jiangxi Agricultural University, Nanchang, China
| | - Jinming You
- Jiangxi Province Key Laboratory of Animal Nutrition, Engineering Research Center of Feed Development, Jiangxi Agricultural University, Nanchang, China
| |
Collapse
|
41
|
Ochoa-Rosales C, Portilla-Fernandez E, Nano J, Wilson R, Lehne B, Mishra PP, Gao X, Ghanbari M, Rueda-Ochoa OL, Juvinao-Quintero D, Loh M, Zhang W, Kooner JS, Grabe HJ, Felix SB, Schöttker B, Zhang Y, Gieger C, Müller-Nurasyid M, Heier M, Peters A, Lehtimäki T, Teumer A, Brenner H, Waldenberger M, Ikram MA, van Meurs JBJ, Franco OH, Voortman T, Chambers J, Stricker BH, Muka T. Epigenetic Link Between Statin Therapy and Type 2 Diabetes. Diabetes Care 2020; 43:875-884. [PMID: 32033992 DOI: 10.2337/dc19-1828] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 01/14/2020] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To investigate the role of epigenetics in statins' diabetogenic effect comparing DNA methylation (DNAm) between statin users and nonusers in an epigenome-wide association study in blood. RESEARCH DESIGN AND METHODS Five cohort studies' participants (n = 8,270) were classified as statin users when they were on statin therapy at the time of DNAm assessment with Illumina 450K or EPIC array or noncurrent users otherwise. Associations of DNAm with various outcomes like incident type 2 diabetes, plasma glucose, insulin, and insulin resistance (HOMA of insulin resistance [HOMA-IR]) as well as with gene expression were investigated. RESULTS Discovery (n = 6,820) and replication (n = 1,450) phases associated five DNAm sites with statin use: cg17901584 (1.12 × 10-25 [DHCR24]), cg10177197 (3.94 × 10-08 [DHCR24]), cg06500161 (2.67 × 10-23 [ABCG1]), cg27243685 (6.01 × 10-09 [ABCG1]), and cg05119988 (7.26 × 10-12 [SC4MOL]). Two sites were associated with at least one glycemic trait or type 2 diabetes. Higher cg06500161 methylation was associated with higher fasting glucose, insulin, HOMA-IR, and type 2 diabetes (odds ratio 1.34 [95% CI 1.22, 1.47]). Mediation analyses suggested that ABCG1 methylation partially mediates the effect of statins on high insulin and HOMA-IR. Gene expression analyses showed that statin exposure and ABCG1 methylation were associated with ABCG1 downregulation, suggesting epigenetic regulation of ABCG1 expression. Further, outcomes insulin and HOMA-IR were significantly associated with ABCG1 expression. CONCLUSIONS This study sheds light on potential mechanisms linking statins with type 2 diabetes risk, providing evidence on DNAm partially mediating statins' effects on insulin traits. Further efforts shall disentangle the molecular mechanisms through which statins may induce DNAm changes, potentially leading to ABCG1 epigenetic regulation.
Collapse
Affiliation(s)
- Carolina Ochoa-Rosales
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- Centro de Vida Saludable de la Universidad de Concepción, Concepción, Chile
| | | | - Jana Nano
- Institute of Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Rory Wilson
- Institute of Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
- Research Unit Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Benjamin Lehne
- Department of Epidemiology and Biostatistics, Imperial College London, London, U.K
| | - Pashupati P Mishra
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Xu Gao
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- Department of Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Oscar L Rueda-Ochoa
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- Electrocardiography Research group, School of Medicine, Universidad Industrial de Santander, Bucaramanga, Colombia
| | | | - Marie Loh
- Department of Epidemiology and Biostatistics, Imperial College London, London, U.K
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Weihua Zhang
- Department of Epidemiology and Biostatistics, Imperial College London, London, U.K
- Department of Cardiology, Ealing Hospital, London North West University Healthcare NHS Trust, Middlesex, U.K
| | - Jaspal S Kooner
- Department of Cardiology, Ealing Hospital, London North West University Healthcare NHS Trust, Middlesex, U.K
- National Heart and Lung Institute, Imperial College London, London, U.K
- Imperial College Healthcare NHS Trust, London, U.K
- MRC-PHE Centre for Environment and Health, Imperial College London, London, U.K
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Stephan B Felix
- Partner Site Greifswald, German Center for Cardiovascular Research (DZHK), Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Ben Schöttker
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Network Aging Research, University of Heidelberg, Heidelberg, Germany
| | - Yan Zhang
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christian Gieger
- Institute of Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
- Research Unit Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Martina Müller-Nurasyid
- Institute for Medical Information Processing, Biometry, and Epidemiology (IBE), Faculty of Medicine, Hospital of the Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
- Department of Internal Medicine I (Cardiology), Hospital of the Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Margit Heier
- Institute of Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
- KORA Study Centre, University Hospital of Augsburg, Augsburg, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
- Research Unit Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Alexander Teumer
- Partner Site Greifswald, German Center for Cardiovascular Research (DZHK), Greifswald, Germany
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Network Aging Research, University of Heidelberg, Heidelberg, Germany
| | - Melanie Waldenberger
- Institute of Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
- Research Unit Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Joyce B J van Meurs
- Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Oscar H Franco
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Trudy Voortman
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - John Chambers
- Department of Epidemiology and Biostatistics, Imperial College London, London, U.K
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Cardiology, Ealing Hospital, London North West University Healthcare NHS Trust, Middlesex, U.K
- Imperial College Healthcare NHS Trust, London, U.K
- MRC-PHE Centre for Environment and Health, Imperial College London, London, U.K
| | - Bruno H Stricker
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Taulant Muka
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| |
Collapse
|
42
|
Cai Z, Yu C, Fu D, Pan Y, Huang J, Rong Y, Deng L, Chen J, Chen M. Differential metabolic and hepatic transcriptome responses of two miniature pig breeds to high dietary cholesterol. Life Sci 2020; 250:117514. [PMID: 32145306 DOI: 10.1016/j.lfs.2020.117514] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/26/2020] [Accepted: 03/03/2020] [Indexed: 12/12/2022]
Abstract
AIMS Pigs are increasingly used as human metabolic disease models; however, there is insufficient research on breed-related genetic background differences. This study aimed to investigate the differential metabolic responses to high-fat and high-cholesterol (HFC) diet-induced non-alcoholic fatty liver disease (NAFLD) of two miniature pig breeds and explore the molecular mechanisms involved. MAIN METHODS Male Wuzhishan (WZSP) and Tibetan pigs (TP) were randomly fed either a standard or an HFC diet for 24 weeks. Weight, serum lipids, bile acid, insulin resistance, liver function, liver histology, and hepatic lipid deposition were determined. RNA-Seq was used to detect the hepatic gene expression profiles. Western blot, immunohistochemistry, and qRT-PCR were used to detect the lipid and glucose metabolism-related gene expressions. KEY FINDINGS The HFC diet caused obesity, hypertension, severe hypercholesterolemia, liver injury, increased hepatocellular steatosis and inflammation, and significantly increased serum insulin levels in both pig breeds. This diet led to higher serum and hepatic cholesterol level concentrations in WZSP and elevated fasting glucose levels in TP. Transcriptome analysis revealed that the genes controlling hepatic cholesterol metabolism and the inflammatory response were consistently regulated; lipid metabolism and insulin signaling related genes were uniquely regulated by the HFC diet in the WZSP and TP, respectively. SIGNIFICANCE Our study demonstrated that the genetic background affects profoundly pigs' metabolic and hepatic responses to an HFC diet. These results deepened our understanding of the molecular mechanisms of HFC diet-induced NAFLD and provided a foundation for selecting the appropriate pig breeds for metabolic studies in the future.
Collapse
Affiliation(s)
- Zhaowei Cai
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Institute of Comparative Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Chen Yu
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Institute of Comparative Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Danting Fu
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Institute of Comparative Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yongming Pan
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Institute of Comparative Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Junjie Huang
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Institute of Comparative Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yili Rong
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Institute of Comparative Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Liqun Deng
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Institute of Comparative Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jiaojiao Chen
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Institute of Comparative Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Minli Chen
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Institute of Comparative Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
43
|
Hafiane A, Favari E, Daskalopoulou SS, Vuilleumier N, Frias MA. High-density lipoprotein cholesterol efflux capacity and cardiovascular risk in autoimmune and non-autoimmune diseases. Metabolism 2020; 104:154141. [PMID: 31923386 DOI: 10.1016/j.metabol.2020.154141] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/23/2019] [Accepted: 01/05/2020] [Indexed: 12/22/2022]
Abstract
Functional assessment of cholesterol efflux capacity (CEC) to high-density lipoprotein (HDL) is an emerging tool for evaluating morbidity and mortality associated with cardiovascular disease (CVD). By promoting macrophage reverse cholesterol transport (RCT), HDL-mediated CEC is believed to play an important role in atherosclerotic lesion progression in the vessel wall. Furthermore, recent evidence indicates that the typical inverse associations between various forms of CEC and CV events may be strongly modulated by environmental systemic factors and traditional CV risk factors, in addition to autoimmune diseases. These factors influence the complex and dynamic composition of HDL particles, which in turn positively or negatively affect HDL-CEC. Herein, we review recent findings connecting HDL-CEC to traditional CV risk factors and cardiometabolic conditions (non-autoimmune diseases) as well as autoimmune diseases, with a specific focus on how these factors may influence the associations between HDL-CEC and CVD risk.
Collapse
Affiliation(s)
- Anouar Hafiane
- Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University, 1001 Decarie Blvd, Bloc E01. 3370H, Montréal, Qc H4A 3J1, Canada.
| | - Elda Favari
- Department of Food and Drug, University of Parma, Parco Area delle Scienze, 27/A, 43124 Parma, Italy.
| | - Stella S Daskalopoulou
- Department of Medicine, Division of Internal Medicine, McGill University, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, EM1.2230, Montreal, Quebec H4A 3J1, Canada.
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine, Diagnostic Department, Geneva University Hospitals, 1211 Geneva, Switzerland; Division of Laboratory Medicine, Department of Medical Specialties, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland.
| | - Miguel A Frias
- Division of Laboratory Medicine, Diagnostic Department, Geneva University Hospitals, 1211 Geneva, Switzerland; Division of Laboratory Medicine, Department of Medical Specialties, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland.
| |
Collapse
|
44
|
Raspberry supplementation reduces lipid accumulation and improves insulin sensitivity in skeletal muscle of mice fed a high-fat diet. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.103572] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
45
|
Ménégaut L, Jalil A, Thomas C, Masson D. Macrophage fatty acid metabolism and atherosclerosis: The rise of PUFAs. Atherosclerosis 2019; 291:52-61. [PMID: 31693943 DOI: 10.1016/j.atherosclerosis.2019.10.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 09/26/2019] [Accepted: 10/09/2019] [Indexed: 01/24/2023]
Abstract
Among the pathways involved in the regulation of macrophage functions, the metabolism of unsaturated fatty acids is central. Indeed, unsaturated fatty acids act as precursors of bioactive molecules such as prostaglandins, leukotrienes, resolvins and related compounds. As components of phospholipids, they have a pivotal role in cell biology by regulating membrane fluidity and membrane-associated cellular processes. Finally, polyunsaturated fatty acids (PUFAs) are also endowed with ligand properties for numerous membrane or nuclear receptors. Although myeloid cells are dependent on the metabolic context for the uptake of essential FAs, recent studies showed that these cells autonomously handle the synthesis of n-3 and n-6 long chain PUFAs such as arachidonic acid and eicosapentaenoic acid. Moreover, targeting PUFA metabolism in macrophages influences pathological processes, including atherosclerosis, by modulating macrophage functions. Omics evidence also supports a role for macrophage PUFA metabolism in the development of cardiometabolic diseases in humans. Currently, there is a renewed interest in the role of n-3/n-6 PUFAs and their oxygenated derivatives in the onset of atherosclerosis and plaque rupture. Purified n-3 FA supplementation appears as a potential strategy in the treatment and prevention of cardiovascular diseases. In this context, the ability of immune cells to handle and to synthesize very long chain PUFA must absolutely be integrated and better understood.
Collapse
Affiliation(s)
- Louise Ménégaut
- Univ. Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000, Dijon, France
| | - Antoine Jalil
- Univ. Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000, Dijon, France
| | - Charles Thomas
- Univ. Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000, Dijon, France
| | - David Masson
- Univ. Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, F-21000, Dijon, France.
| |
Collapse
|
46
|
Grice BA, Barton KJ, Covert JD, Kreilach AM, Tackett L, Brozinick JT, Elmendorf JS. Excess membrane cholesterol is an early contributing reversible aspect of skeletal muscle insulin resistance in C57BL/6NJ mice fed a Western-style high-fat diet. Am J Physiol Endocrinol Metab 2019; 317:E362-E373. [PMID: 31237447 PMCID: PMC6732462 DOI: 10.1152/ajpendo.00396.2018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Skeletal muscle insulin resistance manifests shortly after high-fat feeding, yet mechanisms are not known. Here we set out to determine whether excess skeletal muscle membrane cholesterol and cytoskeletal derangement known to compromise glucose transporter (GLUT)4 regulation occurs early after high-fat feeding. We fed 6-wk-old male C57BL/6NJ mice either a low-fat (LF, 10% kcal) or a high-fat (HF, 45% kcal) diet for 1 wk. This HF feeding challenge was associated with an increase, albeit slight, in body mass, glucose intolerance, and hyperinsulinemia. Liver analyses did not reveal signs of hepatic insulin resistance; however, skeletal muscle immunoblots of triad-enriched regions containing transverse tubule membrane showed a marked loss of stimulated GLUT4 recruitment. An increase in cholesterol was also found in these fractions from HF-fed mice. These derangements were associated with a marked loss of cortical filamentous actin (F-actin) that is essential for GLUT4 regulation and known to be compromised by increases in membrane cholesterol. Both the withdrawal of the HF diet and two subcutaneous injections of the cholesterol-lowering agent methyl-β-cyclodextrin at 3 and 6 days during the 1-wk HF feeding intervention completely mitigated cholesterol accumulation, cortical F-actin loss, and GLUT4 dysregulation. Moreover, these beneficial membrane/cytoskeletal changes occurred concomitant with a full restoration of metabolic responses. These results identify skeletal muscle membrane cholesterol accumulation as an early, reversible, feature of insulin resistance and suggest cortical F-actin loss as an early derangement of skeletal muscle insulin resistance.
Collapse
Affiliation(s)
- Brian A Grice
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, Indiana
| | - Kelly J Barton
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jacob D Covert
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, Indiana
| | - Alec M Kreilach
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, Indiana
| | - Lixuan Tackett
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, Indiana
| | - Joseph T Brozinick
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana
- Eli Lilly and Company, Indianapolis, Indiana
| | - Jeffrey S Elmendorf
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
47
|
Prioritizing complex disease risk genes by integrating multiple data. Genomics 2019; 111:590-597. [DOI: 10.1016/j.ygeno.2018.03.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 03/07/2018] [Accepted: 03/18/2018] [Indexed: 01/18/2023]
|
48
|
Liu J, Carnero-Montoro E, van Dongen J, Lent S, Nedeljkovic I, Ligthart S, Tsai PC, Martin TC, Mandaviya PR, Jansen R, Peters MJ, Duijts L, Jaddoe VWV, Tiemeier H, Felix JF, Willemsen G, de Geus EJC, Chu AY, Levy D, Hwang SJ, Bressler J, Gondalia R, Salfati EL, Herder C, Hidalgo BA, Tanaka T, Moore AZ, Lemaitre RN, Jhun MA, Smith JA, Sotoodehnia N, Bandinelli S, Ferrucci L, Arnett DK, Grallert H, Assimes TL, Hou L, Baccarelli A, Whitsel EA, van Dijk KW, Amin N, Uitterlinden AG, Sijbrands EJG, Franco OH, Dehghan A, Spector TD, Dupuis J, Hivert MF, Rotter JI, Meigs JB, Pankow JS, van Meurs JBJ, Isaacs A, Boomsma DI, Bell JT, Demirkan A, van Duijn CM. An integrative cross-omics analysis of DNA methylation sites of glucose and insulin homeostasis. Nat Commun 2019; 10:2581. [PMID: 31197173 PMCID: PMC6565679 DOI: 10.1038/s41467-019-10487-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 05/09/2019] [Indexed: 02/07/2023] Open
Abstract
Despite existing reports on differential DNA methylation in type 2 diabetes (T2D) and obesity, our understanding of its functional relevance remains limited. Here we show the effect of differential methylation in the early phases of T2D pathology by a blood-based epigenome-wide association study of 4808 non-diabetic Europeans in the discovery phase and 11,750 individuals in the replication. We identify CpGs in LETM1, RBM20, IRS2, MAN2A2 and the 1q25.3 region associated with fasting insulin, and in FCRL6, SLAMF1, APOBEC3H and the 15q26.1 region with fasting glucose. In silico cross-omics analyses highlight the role of differential methylation in the crosstalk between the adaptive immune system and glucose homeostasis. The differential methylation explains at least 16.9% of the association between obesity and insulin. Our study sheds light on the biological interactions between genetic variants driving differential methylation and gene expression in the early pathogenesis of T2D.
Collapse
Affiliation(s)
- Jun Liu
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015GD, The Netherlands. .,Nuffield Department of Population Health, University of Oxford, Oxford, OX3 7FL, UK.
| | - Elena Carnero-Montoro
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015GD, The Netherlands.,Center for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Government, PTS, Granada, 18007, Spain.,Department of Twin Research and Genetic Epidemiology, King's College London, London, WC2R 2LS, UK
| | - Jenny van Dongen
- Department of Biological Psychology, Amsterdam Public Health (APH) research institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, 1081BT, The Netherlands
| | - Samantha Lent
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Ivana Nedeljkovic
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015GD, The Netherlands
| | - Symen Ligthart
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015GD, The Netherlands
| | - Pei-Chien Tsai
- Department of Twin Research and Genetic Epidemiology, King's College London, London, WC2R 2LS, UK.,Department of Biomedical Sciences, Chang Gung University, Taoyuan, 333, Taiwan.,Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, 333, Taiwan
| | - Tiphaine C Martin
- Department of Twin Research and Genetic Epidemiology, King's College London, London, WC2R 2LS, UK.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Pooja R Mandaviya
- Department of Internal Medicine, Section of Pharmacology Vascular and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, 3015GD, The Netherlands
| | - Rick Jansen
- Department of Psychiatry and Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, 1081BT, The Netherlands
| | - Marjolein J Peters
- Department of Internal Medicine, Section of Pharmacology Vascular and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, 3015GD, The Netherlands
| | - Liesbeth Duijts
- Division of Neonatology, Department of Pediatrics, Erasmus University Medical Center, Rotterdam, 3015GD, The Netherlands.,Division of Respiratory Medicine, Department of Pediatrics, Erasmus University Medical Center, Rotterdam, 3015GD, The Netherlands
| | - Vincent W V Jaddoe
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015GD, The Netherlands.,Department of Pediatrics, Erasmus University Medical Center, Rotterdam, 3015GD, The Netherlands.,Generation R Study Group, Erasmus University Medical Center, Rotterdam, 3015GD, The Netherlands
| | - Henning Tiemeier
- Department of Child and Adolescent Psychiatry, Erasmus University Medical Center, Rotterdam, 3015GD, The Netherlands.,Department of Social and Behavioral Science, Harvard TH Chan School of Public Health, Boston, MA, 02115, USA
| | - Janine F Felix
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015GD, The Netherlands.,Department of Pediatrics, Erasmus University Medical Center, Rotterdam, 3015GD, The Netherlands.,Generation R Study Group, Erasmus University Medical Center, Rotterdam, 3015GD, The Netherlands
| | - Gonneke Willemsen
- Department of Biological Psychology, Amsterdam Public Health (APH) research institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, 1081BT, The Netherlands
| | - Eco J C de Geus
- Department of Biological Psychology, Amsterdam Public Health (APH) research institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, 1081BT, The Netherlands
| | - Audrey Y Chu
- The Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20814, USA.,The Framingham Heart Study, National Heart, Lung and Blood Institute, National Institutes of Health, Framingham, MA, 01702, USA
| | - Daniel Levy
- The Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20814, USA.,The Framingham Heart Study, National Heart, Lung and Blood Institute, National Institutes of Health, Framingham, MA, 01702, USA
| | - Shih-Jen Hwang
- The Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20814, USA.,The Framingham Heart Study, National Heart, Lung and Blood Institute, National Institutes of Health, Framingham, MA, 01702, USA
| | - Jan Bressler
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Rahul Gondalia
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Elias L Salfati
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Christian Herder
- German Center for Diabetes Research (DZD), München-Neuherberg, 85764, Germany.,Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, 40225, Germany.,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, 40225, Germany
| | - Bertha A Hidalgo
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Toshiko Tanaka
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, 21224, USA
| | - Ann Zenobia Moore
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, 21224, USA
| | - Rozenn N Lemaitre
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, 98101, USA
| | - Min A Jhun
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, 98101, USA
| | | | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, 21224, USA
| | - Donna K Arnett
- School of Public Health, University of Kentucky, Lexington, KY, 40536, USA
| | - Harald Grallert
- German Center for Diabetes Research (DZD), München-Neuherberg, 85764, Germany.,Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - Themistocles L Assimes
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Lifang Hou
- Center for Population Epigenetics, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University Chicago, Evanston, IL, 60611, USA.,Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Andrea Baccarelli
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, 10032, USA
| | - Eric A Whitsel
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, 27599, USA.,Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina, NC, 27516, USA
| | - Ko Willems van Dijk
- Department of Human Genetics, Leiden University Medical Center, Leiden, 2333ZA, The Netherlands.,Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, 2333ZA, The Netherlands
| | - Najaf Amin
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015GD, The Netherlands
| | - André G Uitterlinden
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015GD, The Netherlands.,Department of Internal Medicine, Section of Pharmacology Vascular and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, 3015GD, The Netherlands
| | - Eric J G Sijbrands
- Department of Internal Medicine, Section of Pharmacology Vascular and Metabolic Diseases, Erasmus University Medical Center, Rotterdam, 3015GD, The Netherlands
| | - Oscar H Franco
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015GD, The Netherlands.,Institute of Social and Preventive Medicine (ISPM), University of Bern, Bern, 3012, Switzerland
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015GD, The Netherlands.,Department of Epidemiology and Biostatistics, Imperial College London, London, SW7 2AZ, UK
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, WC2R 2LS, UK
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Marie-France Hivert
- Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, J1K0A5, Canada.,Diabetes Unit, Massachusetts General Hospital, Boston, MA, 02114, USA.,Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, 02215, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences and Departments of Pediatrics and Medicine, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| | - James B Meigs
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA.,Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA.,Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - James S Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Joyce B J van Meurs
- CARIM School for Cardiovascular Diseases, Maastricht Centre for Systems Biology (MaCSBio), and Departments of Biochemistry and Physiology, Maastricht University, Maastricht, 6211LK, The Netherlands
| | - Aaron Isaacs
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015GD, The Netherlands.,CARIM School for Cardiovascular Diseases, Maastricht Centre for Systems Biology (MaCSBio), and Departments of Biochemistry and Physiology, Maastricht University, Maastricht, 6211LK, The Netherlands
| | - Dorret I Boomsma
- Department of Biological Psychology, Amsterdam Public Health (APH) research institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, 1081BT, The Netherlands
| | - Jordana T Bell
- Department of Twin Research and Genetic Epidemiology, King's College London, London, WC2R 2LS, UK
| | - Ayşe Demirkan
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015GD, The Netherlands. .,Department of Genetics, University Medical Center Groningen, Groningen, 9713GZ, The Netherlands. .,Section of Statistical Multi-Omics, Department of Experimental and Clinical Research, School of Bioscience and Medicine, Univeristy of Surrey, Guildford, GU2 7XH, UK.
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015GD, The Netherlands. .,Nuffield Department of Population Health, University of Oxford, Oxford, OX3 7FL, UK. .,Leiden Academic Center for Drug Research, Leiden University, Leiden, 2311EZ, The Netherlands.
| |
Collapse
|
49
|
Ma J, Nano J, Ding J, Zheng Y, Hennein R, Liu C, Speliotes EK, Huan T, Song C, Mendelson MM, Joehanes R, Long MT, Liang L, Smith JA, Reynolds LM, Ghanbari M, Muka T, van Meurs JBJ, Alferink LJM, Franco OH, Dehghan A, Ratliff S, Zhao W, Bielak L, Kardia SLR, Peyser PA, Ning H, VanWagner LB, Lloyd-Jones DM, Carr JJ, Greenland P, Lichtenstein AH, Hu FB, Liu Y, Hou L, Darwish Murad S, Levy D. A Peripheral Blood DNA Methylation Signature of Hepatic Fat Reveals a Potential Causal Pathway for Nonalcoholic Fatty Liver Disease. Diabetes 2019; 68:1073-1083. [PMID: 30936141 PMCID: PMC6477898 DOI: 10.2337/db18-1193] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 02/14/2019] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a risk factor for type 2 diabetes (T2D). We aimed to identify the peripheral blood DNA methylation signature of hepatic fat. We conducted epigenome-wide association studies of hepatic fat in 3,400 European ancestry (EA) participants and in 401 Hispanic ancestry and 724 African ancestry participants from four population-based cohort studies. Hepatic fat was measured using computed tomography or ultrasound imaging and DNA methylation was assessed at >400,000 cytosine-guanine dinucleotides (CpGs) in whole blood or CD14+ monocytes using a commercial array. We identified 22 CpGs associated with hepatic fat in EA participants at a false discovery rate <0.05 (corresponding P = 6.9 × 10-6) with replication at Bonferroni-corrected P < 8.6 × 10-4 Mendelian randomization analyses supported the association of hypomethylation of cg08309687 (LINC00649) with NAFLD (P = 2.5 × 10-4). Hypomethylation of the same CpG was also associated with risk for new-onset T2D (P = 0.005). Our study demonstrates that a peripheral blood-derived DNA methylation signature is robustly associated with hepatic fat accumulation. The hepatic fat-associated CpGs may represent attractive biomarkers for T2D. Future studies are warranted to explore mechanisms and to examine DNA methylation signatures of NAFLD across racial/ethnic groups.
Collapse
Affiliation(s)
- Jiantao Ma
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, and Framingham Heart Study, Framingham, MA
- Nutrition Data Science, Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA
| | - Jana Nano
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Epidemiology, Neuherberg, Germany
- German Center for Diabetes Research, München-Neuherberg, Germany
| | - Jingzhong Ding
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC
| | - Yinan Zheng
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Rachel Hennein
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, and Framingham Heart Study, Framingham, MA
| | - Chunyu Liu
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, and Framingham Heart Study, Framingham, MA
- Department of Biostatistics, Boston University, Boston, MA
| | | | - Tianxiao Huan
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, and Framingham Heart Study, Framingham, MA
| | - Ci Song
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, and Framingham Heart Study, Framingham, MA
- Molecular Epidemiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Michael M Mendelson
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, and Framingham Heart Study, Framingham, MA
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Roby Joehanes
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, and Framingham Heart Study, Framingham, MA
| | - Michelle T Long
- Section of Gastroenterology, Department of Medicine, Boston University School of Medicine, Boston, MA
| | - Liming Liang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI
| | - Lindsay M Reynolds
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Taulant Muka
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Joyce B J van Meurs
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Louise J M Alferink
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Oscar H Franco
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Biostatistics and Epidemiology, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, U.K
| | - Scott Ratliff
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI
| | - Lawrence Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI
| | - Patricia A Peyser
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI
| | - Hongyan Ning
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Lisa B VanWagner
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
- Division of Gastroenterology and Hepatology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Donald M Lloyd-Jones
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - John Jeffrey Carr
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN
| | - Philip Greenland
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Alice H Lichtenstein
- Cardiovascular Nutrition Laboratory, USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Yongmei Liu
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC
| | - Lifang Hou
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Sarwa Darwish Murad
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Daniel Levy
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, and Framingham Heart Study, Framingham, MA
| |
Collapse
|
50
|
Tan ALM, Langley SR, Tan CF, Chai JF, Khoo CM, Leow MKS, Khoo EYH, Moreno-Moral A, Pravenec M, Rotival M, Sadananthan SA, Velan SS, Venkataraman K, Chong YS, Lee YS, Sim X, Stunkel W, Liu MH, Tai ES, Petretto E. Ethnicity-Specific Skeletal Muscle Transcriptional Signatures and Their Relevance to Insulin Resistance in Singapore. J Clin Endocrinol Metab 2019; 104:465-486. [PMID: 30137523 DOI: 10.1210/jc.2018-00309] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 08/14/2018] [Indexed: 11/19/2022]
Abstract
CONTEXT Insulin resistance (IR) and obesity differ among ethnic groups in Singapore, with the Malays more obese yet less IR than Asian-Indians. However, the molecular basis underlying these differences is not clear. OBJECTIVE As the skeletal muscle (SM) is metabolically relevant to IR, we investigated molecular pathways in SM that are associated with ethnic differences in IR, obesity, and related traits. DESIGN, SETTING, AND MAIN OUTCOME MEASURES We integrated transcriptomic, genomic, and phenotypic analyses in 156 healthy subjects representing three major ethnicities in the Singapore Adult Metabolism Study. PATIENTS This study contains Chinese (n = 63), Malay (n = 51), and Asian-Indian (n = 42) men, aged 21 to 40 years, without systemic diseases. RESULTS We found remarkable diversity in the SM transcriptome among the three ethnicities, with >8000 differentially expressed genes (40% of all genes expressed in SM). Comparison with blood transcriptome from a separate Singaporean cohort showed that >95% of SM expression differences among ethnicities were unique to SM. We identified a network of 46 genes that were specifically downregulated in Malays, suggesting dysregulation of components of cellular respiration in SM of Malay individuals. We also report 28 differentially expressed gene clusters, four of which were also enriched for genes that were found in genome-wide association studies of metabolic traits and disease and correlated with variation in IR, obesity, and related traits. CONCLUSION We identified extensive gene-expression changes in SM among the three Singaporean ethnicities and report specific genes and molecular pathways that might underpin and explain the differences in IR among these ethnic groups.
Collapse
Affiliation(s)
- Amelia Li Min Tan
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Duke-National University of Singapore Medical School, Singapore
| | - Sarah R Langley
- Duke-National University of Singapore Medical School, Singapore
- National Heart Centre Singapore, Singapore
| | - Chee Fan Tan
- Nanyang Institute of Technology in Health and Medicine, Nanyang Technological University, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Jin Fang Chai
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Chin Meng Khoo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Duke-National University of Singapore Medical School, Singapore
- Division of Endocrinology, Department of Medicine, National University Health System, Singapore
| | - Melvin Khee-Shing Leow
- Duke-National University of Singapore Medical School, Singapore
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Eric Yin Hao Khoo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Division of Endocrinology, Department of Medicine, National University Health System, Singapore
| | | | - Michal Pravenec
- Institute Of Physiology, Czech Academy Of Sciences, Prague, Czech Republic
| | - Maxime Rotival
- Unit of Human Evolutionary Genetics, Institut Pasteur, Paris, France
| | - Suresh Anand Sadananthan
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore
| | - S Sendhil Velan
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Kavita Venkataraman
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Yap Seng Chong
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yung Seng Lee
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Division of Paediatrics Endocrinology, Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, National University Health System, Singapore
| | - Xueling Sim
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Walter Stunkel
- Experimental Biotherapeutics Centre, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Mei Hui Liu
- Department of Chemistry, Food Science & Technology Programme, National University of Singapore, Singapore
| | - E Shyong Tai
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Duke-National University of Singapore Medical School, Singapore
- Division of Endocrinology, Department of Medicine, National University Health System, Singapore
| | - Enrico Petretto
- Duke-National University of Singapore Medical School, Singapore
| |
Collapse
|